1
|
Wuyts FL, Deblieck C, Vandevoorde C, Durante M. Brains in space: impact of microgravity and cosmic radiation on the CNS during space exploration. Nat Rev Neurosci 2025:10.1038/s41583-025-00923-4. [PMID: 40247135 DOI: 10.1038/s41583-025-00923-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2025] [Indexed: 04/19/2025]
Abstract
Solar system exploration is a grand endeavour of humankind. Space agencies have been planning crewed missions to the Moon and Mars for several decades. However, several environmental stress factors in space, such as microgravity and cosmic radiation, confer health risks for human explorers. This Review examines the effects of microgravity and exposure to cosmic radiation on the CNS. Microgravity presents challenges for the brain, necessitating the development of adaptive movement and orientation strategies to cope with alterations in sensory information. Exposure to microgravity also affects cognitive function to a certain extent. Recent MRI results show that microgravity affects brain structure and function. Post-flight recovery from these changes is gradual, with some lasting up to a year. Regarding cosmic radiation, animal experiments suggest that the brain could be much more sensitive to this stressor than may be expected from experiences on Earth. This may be due to the presence of energetic heavy ions in space that have an impact on cognitive function, even at low doses. However, all data about space radiation risk stem from rodent experiments, and extrapolation of these data to humans carries a high degree of uncertainty. Here, after presenting an overview of current knowledge in the above areas, we provide a concise description of possible counter-measures to protect the brain against microgravity and cosmic radiation during future space missions.
Collapse
Affiliation(s)
- Floris L Wuyts
- Laboratory for Equilibrium Investigations and Aerospace (LEIA), University of Antwerp, Antwerp, Belgium
| | - Choi Deblieck
- Laboratory for Equilibrium Investigations and Aerospace (LEIA), University of Antwerp, Antwerp, Belgium
| | - Charlot Vandevoorde
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Marco Durante
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany.
- Institute for Condensed Matter of Physics, Technische Universität Darmstadt, Darmstadt, Germany.
- Department of Physics 'Ettore Pancini', University Federico II, Naples, Italy.
| |
Collapse
|
2
|
Zhang D, Thomas R, Lam TT, Veselinovic I, Grosshans DR. Cranial radiation disrupts dopaminergic signaling and connectivity in the mammalian brain. Acta Neuropathol Commun 2025; 13:59. [PMID: 40083022 PMCID: PMC11905640 DOI: 10.1186/s40478-025-01976-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 02/28/2025] [Indexed: 03/16/2025] Open
Abstract
Cognitive impairment is a common and challenging side effect of cranial radiation therapy for brain tumors, though its precise mechanisms remain unclear. The mesocortical dopaminergic pathway, known to play a key role in cognitive function, is implicated in several neuropsychiatric disorders, yet its involvement in radiation-induced cognitive dysfunction is unexplored. Here, with using in vivo multi-electrode array recordings of both anesthetized and free-moving rats to monitor the firing activities of dopamine neurons in the ventral tegmental area (VTA) and local field potentials in both the prefrontal cortex (PFC) and VTA, as well as the immunofluorescence assays and western blotting, we report that cranial irradiation transiently altered VTA dopamine neuron firing patterns without affecting overall firing rates and led to sustained reductions in both "awake" and total dopamine neuron density. Additionally, radiation exposure impaired D2 receptor function and disrupted connectivity between the PFC and VTA. These multifaceted disruptions in the mesocortical dopamine signaling may underlie the development of radiation-induced cognitive dysfunction. These findings pave the way for novel research to prevent or reverse radiation-induced injury, ultimately improving the quality of life for brain tumor survivors.
Collapse
Affiliation(s)
- Die Zhang
- Departments of Radiation Oncology and Experimental Radiation Oncology, Unit 97, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, 77030-4009, Houston, TX, USA
| | - Riya Thomas
- Departments of Radiation Oncology and Experimental Radiation Oncology, Unit 97, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, 77030-4009, Houston, TX, USA
| | - Thanh Thai Lam
- Departments of Radiation Oncology and Experimental Radiation Oncology, Unit 97, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, 77030-4009, Houston, TX, USA
| | - Ines Veselinovic
- Departments of Radiation Oncology and Experimental Radiation Oncology, Unit 97, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, 77030-4009, Houston, TX, USA
| | - David R Grosshans
- Departments of Radiation Oncology and Experimental Radiation Oncology, Unit 97, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, 77030-4009, Houston, TX, USA.
| |
Collapse
|
3
|
Smits E, Reid FE, Tamgue EN, Alvarado Arriaga P, Nguyen C, Britten RA. Sex-Dependent Changes in Risk-Taking Predisposition of Rats Following Space Radiation Exposure. Life (Basel) 2025; 15:449. [PMID: 40141792 PMCID: PMC11943666 DOI: 10.3390/life15030449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/21/2025] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
The Artemis missions will establish a sustainable human presence on the Moon, serving as a crucial steppingstone for future Mars exploration. Astronauts on these ambitious missions will have to successfully complete complex tasks, which will frequently involve rapid and effective decision making under unfamiliar or high-pressure conditions. Exposure to low doses of space radiation (SR) can impair key executive functions critical to decision making. This study examined the effects of exposure to 10 cGy of Galactic Cosmic Ray simulated radiation (GCRsim) on decision-making performance in male and female rats with a naturally low predisposition for risk-taking (RTP) prior to exposure. Rats were assessed at monthly intervals following SR exposure and the RTP performance contrasted with that observed during the prescreening process. Exposure to 10 cGy of GCRsim impaired decision making in both male and female rats, with sex-dependent outcomes. By 30 days after SR exposure, female rats became more risk-prone, making less profitable decisions, while male rats retained their decision-making strategies but took significantly longer to make selections. However, continued practice in the RTP tasks appeared to reduce/reverse these performance deficits. This study has expanded our understanding of the range of cognitive processes impacted by SR to include decision making.
Collapse
Affiliation(s)
- Elliot Smits
- EVMS School of Medicine, Macon and Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA; (E.S.); (F.E.R.)
| | - Faith E. Reid
- EVMS School of Medicine, Macon and Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA; (E.S.); (F.E.R.)
| | - Ella N. Tamgue
- EVMS Radiation Oncology, Eastern Virginia Medical School, Macon and Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA (P.A.A.); (C.N.)
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Macon and Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA
| | - Paola Alvarado Arriaga
- EVMS Radiation Oncology, Eastern Virginia Medical School, Macon and Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA (P.A.A.); (C.N.)
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Macon and Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA
| | - Charles Nguyen
- EVMS Radiation Oncology, Eastern Virginia Medical School, Macon and Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA (P.A.A.); (C.N.)
| | - Richard A. Britten
- EVMS Radiation Oncology, Eastern Virginia Medical School, Macon and Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA (P.A.A.); (C.N.)
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Macon and Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA
| |
Collapse
|
4
|
Yun S, Kiffer FC, Bancroft GL, Guzman CS, Soler I, Haas HA, Shi R, Patel R, Lara-Jiménez J, Kumar PL, Tran FH, Ahn KJ, Rong Y, Luitel K, Shay JW, Eisch AJ. The longitudinal behavioral effects of acute exposure to galactic cosmic radiation in female C57BL/6J mice: Implications for deep space missions, female crews, and potential antioxidant countermeasures. J Neurochem 2025; 169:e16225. [PMID: 39318241 DOI: 10.1111/jnc.16225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 08/07/2024] [Accepted: 08/24/2024] [Indexed: 09/26/2024]
Abstract
Galactic cosmic radiation (GCR) is an unavoidable risk to astronauts that may affect mission success. Male rodents exposed to 33-beam-GCR (33-GCR) show short-term cognitive deficits but reports on female rodents and long-term assessment are lacking. We asked: What are the longitudinal behavioral effects of 33-GCR on female mice? Also, can an antioxidant/anti-inflammatory compound (CDDO-EA) mitigate the impact of 33-GCR? Mature (6-month-old) C57BL/6J female mice received CDDO-EA (400 μg/g of food) or a control diet (vehicle, Veh) for 5 days and Sham-irradiation (IRR) or whole-body 33-GCR (0.75Gy) on the 4th day. Three-months post-IRR, mice underwent two touchscreen-platform tests: (1) location discrimination reversal (tests behavior pattern separation and cognitive flexibility, abilities reliant on the dentate gyrus) and (2) stimulus-response learning/extinction. Mice then underwent arena-based behavior tests (e.g. open field, 3-chamber social interaction). At the experiment's end (14.25-month post-IRR), an index relevant to neurogenesis was quantified (doublecortin-immunoreactive [DCX+] dentate gyrus immature neurons). Female mice exposed to Veh/Sham vs. Veh/33-GCR had similar pattern separation (% correct to 1st reversal). There were two effects of diet: CDDO-EA/Sham and CDDO-EA/33-GCR mice had better pattern separation vs. their respective control groups (Veh/Sham, Veh/33-GCR), and CDDO-EA/33-GCR mice had better cognitive flexibility (reversal number) vs. Veh/33-GCR mice. One radiation effect/CDDO-EA countereffect also emerged: Veh/33-GCR mice had slower stimulus-response learning (days to completion) vs. all other groups, including CDDO-EA/33-GCR mice. In general, all mice showed normal anxiety-like behavior, exploration, and habituation to novel environments. There was also a change relevant to neurogenesis: Veh/33-GCR mice had fewer DCX+ dentate gyrus immature neurons vs. Veh/Sham mice. Our study implies space radiation is a risk to a female crew's longitudinal mission-relevant cognitive processes and CDDO-EA is a potential dietary countermeasure for space-radiation CNS risks.
Collapse
Affiliation(s)
- Sanghee Yun
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania, USA
- Neuroscience Graduate Group, The University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Frederico C Kiffer
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania, USA
- The University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Grace L Bancroft
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania, USA
- School of Arts and Sciences, The University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Caterina S Guzman
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania, USA
- The University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Ivan Soler
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania, USA
- The University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Harley A Haas
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania, USA
- School of Arts and Sciences, The University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Raymon Shi
- School of Arts and Sciences, The University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Riya Patel
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania, USA
| | - Jaysen Lara-Jiménez
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania, USA
- The University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Priya L Kumar
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania, USA
- School of Arts and Sciences, The University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Fionya H Tran
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania, USA
| | - Kyung Jin Ahn
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania, USA
| | - Yuying Rong
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania, USA
| | - Krishna Luitel
- Department of Cell Biology, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jerry W Shay
- Department of Cell Biology, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Amelia J Eisch
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania, USA
- Neuroscience Graduate Group, The University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Department of Neuroscience, The University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
5
|
Sleiman A, Miller KB, Flores D, Kuan J, Altwasser K, Smith BJ, Kozbenko T, Hocking R, Wood SJ, Huff J, Adam-Guillermin C, Hamada N, Yauk C, Wilkins R, Chauhan V. AOP report: Development of an adverse outcome pathway for deposition of energy leading to learning and memory impairment. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2024; 65 Suppl 3:57-84. [PMID: 39228295 DOI: 10.1002/em.22622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 09/05/2024]
Abstract
Understanding radiation-induced non-cancer effects on the central nervous system (CNS) is essential for the risk assessment of medical (e.g., radiotherapy) and occupational (e.g., nuclear workers and astronauts) exposures. Herein, the adverse outcome pathway (AOP) approach was used to consolidate relevant studies in the area of cognitive decline for identification of research gaps, countermeasure development, and for eventual use in risk assessments. AOPs are an analytical construct describing critical events to an adverse outcome (AO) in a simplified form beginning with a molecular initiating event (MIE). An AOP was constructed utilizing mechanistic information to build empirical support for the key event relationships (KERs) between the MIE of deposition of energy to the AO of learning and memory impairment through multiple key events (KEs). The evidence for the AOP was acquired through a documented scoping review of the literature. In this AOP, the MIE is connected to the AO via six KEs: increased oxidative stress, increased deoxyribonucleic acid (DNA) strand breaks, altered stress response signaling, tissue resident cell activation, increased pro-inflammatory mediators, and abnormal neural remodeling that encompasses atypical structural and functional alterations of neural cells and surrounding environment. Deposition of energy directly leads to oxidative stress, increased DNA strand breaks, an increase of pro-inflammatory mediators and tissue resident cell activation. These KEs, which are themselves interconnected, can lead to abnormal neural remodeling impacting learning and memory processes. Identified knowledge gaps include improving quantitative understanding of the AOP across several KERs and additional testing of proposed modulating factors through experimental work. Broadly, it is envisioned that the outcome of these efforts could be extended to other cognitive disorders and complement ongoing work by international radiation governing bodies in their review of the system of radiological protection.
Collapse
Affiliation(s)
- Ahmad Sleiman
- Institut de Radioprotection et de Sûreté Nucléaire, St. Paul Lez Durance, Provence, France
| | - Kathleen B Miller
- Department of Health and Exercise Science, Morrison College Family of Health, University of St. Thomas, Saint Paul, Minnesota, USA
| | - Danicia Flores
- Consumer and Clinical Radiation Protection Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Ontario, Canada
| | - Jaqueline Kuan
- Consumer and Clinical Radiation Protection Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Ontario, Canada
| | - Kaitlyn Altwasser
- Consumer and Clinical Radiation Protection Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Ontario, Canada
| | - Benjamin J Smith
- Consumer and Clinical Radiation Protection Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Ontario, Canada
| | - Tatiana Kozbenko
- Consumer and Clinical Radiation Protection Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Ontario, Canada
| | - Robyn Hocking
- Consumer and Clinical Radiation Protection Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Ontario, Canada
| | | | - Janice Huff
- NASA Langley Research Center, Hampton, Virginia, USA
| | | | - Nobuyuki Hamada
- Biology and Environmental Chemistry Division, Sustainable System Research Laboratory, Central Research Institute of Electric Power Industry (CRIEPI), Chiba, Japan
| | - Carole Yauk
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Ruth Wilkins
- Consumer and Clinical Radiation Protection Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Ontario, Canada
| | - Vinita Chauhan
- Consumer and Clinical Radiation Protection Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Ontario, Canada
| |
Collapse
|
6
|
Yun S, Kiffer FC, Bancroft GL, Guzman CS, Soler I, Haas HA, Shi R, Patel R, Lara-Jiménez J, Kumar PL, Tran FH, Ahn KJ, Rong Y, Luitel K, Shay JW, Eisch AJ. The longitudinal behavioral effects of acute exposure to galactic cosmic radiation in female C57BL/6J mice: implications for deep space missions, female crews, and potential antioxidant countermeasures. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.12.588768. [PMID: 38659963 PMCID: PMC11042186 DOI: 10.1101/2024.04.12.588768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Galactic cosmic radiation (GCR) is an unavoidable risk to astronauts that may affect mission success. Male rodents exposed to 33-beam-GCR (33-GCR) show short-term cognitive deficits but reports on female rodents and long-term assessment is lacking. Here we asked: What are the longitudinal behavioral effects of 33-GCR on female mice? Also, can an antioxidant/anti-inflammatory compound mitigate the impact of 33-GCR? Mature (6-month-old) C57BL/6J female mice received the antioxidant CDDO-EA (400 µg/g of food) or a control diet (vehicle, Veh) for 5 days and either Sham-irradiation (IRR) or whole-body 33-GCR (0.75Gy) on the 4th day. Three-months post-IRR, mice underwent two touchscreen-platform tests: 1) location discrimination reversal (which tests behavior pattern separation and cognitive flexibility, two abilities reliant on the dentate gyrus) and 2) stimulus-response learning/extinction. Mice then underwent arena-based behavior tests (e.g. open field, 3-chamber social interaction). At the experiment end (14.25-month post-IRR), neurogenesis was assessed (doublecortin-immunoreactive [DCX+] dentate gyrus neurons). Female mice exposed to Veh/Sham vs. Veh/33-GCR had similar pattern separation (% correct to 1st reversal). There were two effects of diet: CDDO-EA/Sham and CDDO-EA/33-GCR mice had better pattern separation vs. their respective control groups (Veh/Sham, Veh/33-GCR), and CDDO-EA/33-GCR mice had better cognitive flexibility (reversal number) vs. Veh/33-GCR mice. Notably, one radiation effect/CDDO-EA countereffect also emerged: Veh/33-GCR mice had worse stimulus-response learning (days to completion) vs. all other groups, including CDDO-EA/33-GCR mice. In general, all mice show normal anxiety-like behavior, exploration, and habituation to novel environments. There was also a change in neurogenesis: Veh/33-GCR mice had fewer DCX+ dentate gyrus immature neurons vs. Veh/Sham mice. Our study implies space radiation is a risk to a female crew's longitudinal mission-relevant cognitive processes and CDDO-EA is a potential dietary countermeasure for space-radiation CNS risks.
Collapse
|
7
|
Britten RA, Fesshaye A, Tidmore A, Liu A, Blackwell AA. Loss of Cognitive Flexibility Practice Effects in Female Rats Exposed to Simulated Space Radiation. Radiat Res 2023; 200:256-265. [PMID: 37527363 DOI: 10.1667/rade-22-00196.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 06/27/2023] [Indexed: 08/03/2023]
Abstract
During the planned missions to Mars, astronauts will be faced with many potential health hazards including prolonged exposure to space radiation. Ground-based studies have shown that exposure to space radiation impairs the performance of male rats in cognitive flexibility tasks which involve processes that are essential to rapidly and efficiently adapting to different situations. However, there is presently a paucity of information on the effects of space radiation on cognitive flexibility in female rodents. This study has determined the impact that exposure to a low (10 cGy) dose of ions from the simplified 5-ion galactic cosmic ray simulation [https://www.bnl.gov/nsrl/userguide/SimGCRSim.php (07/2023)] (GCRSim) beam or 250 MeV/n 4He ions has on the ability of female Wistar rats to perform in constrained [attentional set shifting (ATSET)] and unconstrained cognitive flexibility (UCFlex) tasks. Female rats exposed to GCRSim exhibited multiple decrements in ATSET performance. Firstly, GCRSim exposure impaired performance in the compound discrimination (CD) stage of the ATSET task. While the ability of rats to identify the rewarded cue was not compromised, the time the rats required to do so significantly increased. Secondly, both 4He and GCRSim exposure reduced the ability of rats to reach criterion in the compound discrimination reversal (CDR) stage. Approximately 20% of the irradiated rats were unable to complete the CDR task; furthermore, the irradiated rats that did reach criterion took more attempts to do so than did the sham-treated animals. Radiation exposure also altered the magnitude and/or nature of practice effects. A comparison of performance metrics from the pre-screen and post-exposure ATSET task revealed that while the sham-treated rats completed the post-exposure CD stage of the ATSET task in 30% less time than for completion of the pre-screen ATSET task, the irradiated rats took 30-50% longer to do so. Similarly, while sham-treated rats completed the CDR stage in ∼10% fewer attempts in the post-exposure task compared to the pre-screen task, in contrast, the 4He- and GCRSim-exposed cohorts took more (∼2-fold) attempts to reach criterion in the post-exposure task than in the pre-screen task. In conclusion, this study demonstrates that female rats are susceptible to radiation-induced loss of performance in the constrained ATSET cognitive flexibility task. Moreover, exposure to radiation leads to multiple performance decrements, including loss of practice effects, an increase in anterograde interference and reduced ability or unwillingness to switch attention. Should similar effects occur in humans, astronauts may have a compromised ability to perform complex tasks.
Collapse
Affiliation(s)
- Richard A Britten
- EVMS Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507
- EVMS Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, Virginia 23507
- Center for Integrative Neuroscience and Inflammatory diseases, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Arriyam Fesshaye
- EVMS Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Alyssa Tidmore
- EVMS Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Aiyi Liu
- EVMS Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Ashley A Blackwell
- EVMS Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507
- Center for Integrative Neuroscience and Inflammatory diseases, Eastern Virginia Medical School, Norfolk, Virginia 23507
| |
Collapse
|
8
|
Kremsky I, Ali S, Stanbouly S, Holley J, Justinen S, Pecaut M, Crapo J, Mao X. Spaceflight-Induced Gene Expression Profiles in the Mouse Brain Are Attenuated by Treatment with the Antioxidant BuOE. Int J Mol Sci 2023; 24:13569. [PMID: 37686374 PMCID: PMC10487739 DOI: 10.3390/ijms241713569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 08/25/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
The demands of deep space pose a health risk to the central nervous system that has long been a concern when sending humans to space. While little is known about how spaceflight affects transcription spatially in the brain, a greater understanding of this process has the potential to aid strategies that mitigate the effects of spaceflight on the brain. Therefore, we performed GeoMx Digital Spatial Profiling of mouse brains subjected to either spaceflight or grounded controls. Four brain regions were selected: Cortex, Frontal Cortex, Corunu Ammonis I, and Dentate Gyrus. Antioxidants have emerged as a potential means of attenuating the effects of spaceflight, so we treated a subset of the mice with a superoxide dismutase mimic, MnTnBuOE-2-PyP 5+ (BuOE). Our analysis revealed hundreds of differentially expressed genes due to spaceflight in each of the four brain regions. Both common and region-specific transcriptomic responses were observed. Metabolic pathways and pathways sensitive to oxidative stress were enriched in the four brain regions due to spaceflight. These findings enhance our understanding of brain regional variation in susceptibility to spaceflight conditions. BuOE reduced the transcriptomic effects of spaceflight at a large number of genes, suggesting that this compound may attenuate oxidative stress-induced brain damage caused by the spaceflight environment.
Collapse
Affiliation(s)
- Isaac Kremsky
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (I.K.); (S.A.); (S.S.); (J.H.); (S.J.); (M.P.)
- Center for Genomics, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Samir Ali
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (I.K.); (S.A.); (S.S.); (J.H.); (S.J.); (M.P.)
| | - Seta Stanbouly
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (I.K.); (S.A.); (S.S.); (J.H.); (S.J.); (M.P.)
| | - Jacob Holley
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (I.K.); (S.A.); (S.S.); (J.H.); (S.J.); (M.P.)
| | - Stephen Justinen
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (I.K.); (S.A.); (S.S.); (J.H.); (S.J.); (M.P.)
| | - Michael Pecaut
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (I.K.); (S.A.); (S.S.); (J.H.); (S.J.); (M.P.)
| | - James Crapo
- Department of Medicine, Division of Pulmonary, Critical Care & Sleep Medicine, National Jewish Health, University of Colorado Denver, Denver, CO 80206, USA;
| | - Xiaowen Mao
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (I.K.); (S.A.); (S.S.); (J.H.); (S.J.); (M.P.)
| |
Collapse
|
9
|
Alwood JS, Mulavara AP, Iyer J, Mhatre SD, Rosi S, Shelhamer M, Davis C, Jones CW, Mao XW, Desai RI, Whitmire AM, Williams TJ. Circuits and Biomarkers of the Central Nervous System Relating to Astronaut Performance: Summary Report for a NASA-Sponsored Technical Interchange Meeting. Life (Basel) 2023; 13:1852. [PMID: 37763256 PMCID: PMC10532466 DOI: 10.3390/life13091852] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/24/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Biomarkers, ranging from molecules to behavior, can be used to identify thresholds beyond which performance of mission tasks may be compromised and could potentially trigger the activation of countermeasures. Identification of homologous brain regions and/or neural circuits related to operational performance may allow for translational studies between species. Three discussion groups were directed to use operationally relevant performance tasks as a driver when identifying biomarkers and brain regions or circuits for selected constructs. Here we summarize small-group discussions in tables of circuits and biomarkers categorized by (a) sensorimotor, (b) behavioral medicine and (c) integrated approaches (e.g., physiological responses). In total, hundreds of biomarkers have been identified and are summarized herein by the respective group leads. We hope the meeting proceedings become a rich resource for NASA's Human Research Program (HRP) and the community of researchers.
Collapse
Affiliation(s)
| | | | - Janani Iyer
- Universities Space Research Association (USRA), Moffett Field, CA 94035, USA
| | | | - Susanna Rosi
- Department of Physical Therapy & Rehabilitation Science, University of California, San Francisco, CA 94110, USA
- Department of Neurological Surgery, University of California, San Francisco, CA 94110, USA
| | - Mark Shelhamer
- Department of Otolaryngology–Head and Neck Surgery, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Catherine Davis
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences (USUHS), Bethesda, MD 20814, USA
| | - Christopher W. Jones
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xiao Wen Mao
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University Health, Loma Linda, CA 92354, USA
| | - Rajeev I. Desai
- Integrative Neurochemistry Laboratory, Behavioral Biology Program, McLean Hospital-Harvard Medical School, Belmont, MA 02478, USA
| | | | | |
Collapse
|
10
|
Sanford LD, Adkins AM, Boden AF, Gotthold JD, Harris RD, Shuboni-Mulligan D, Wellman LL, Britten RA. Sleep and Core Body Temperature Alterations Induced by Space Radiation in Rats. Life (Basel) 2023; 13:life13041002. [PMID: 37109531 PMCID: PMC10144689 DOI: 10.3390/life13041002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/09/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Sleep problems in astronauts can arise from mission demands and stress and can impact both their health and ability to accomplish mission objectives. In addition to mission-related physical and psychological stressors, the long durations of the proposed Mars missions will expose astronauts to space radiation (SR), which has a significant impact on the brain and may also alter sleep and physiological functions. Therefore, in this study, we assessed sleep, EEG spectra, activity, and core body temperature (CBT) in rats exposed to SR and compared them to age-matched nonirradiated rats. Male outbred Wistar rats (8-9 months old at the time of the study) received SR (15 cGy GCRsim, n = 15) or served as age- and time-matched controls (CTRL, n = 15) without irradiation. At least 90 days after SR and 3 weeks prior to recording, all rats were implanted with telemetry transmitters for recording EEG, activity, and CBT. Sleep, EEG spectra (delta, 0.5-4 Hz; theta, 4-8 Hz; alpha, 8-12 Hz; sigma, 12-16 Hz; beta, 16-24 Hz), activity, and CBT were examined during light and dark periods and during waking and sleeping states. When compared to the CTRLs, SR produced significant reductions in the amounts of dark period total sleep time, total nonrapid eye movement sleep (NREM), and total rapid eye movement sleep (REM), with significant decreases in light and dark period NREM deltas and dark period REM thetas as well as increases in alpha and sigma in NREM and REM during either light or dark periods. The SR animals showed modest increases in some measures of activity. CBT was significantly reduced during waking and sleeping in the light period. These data demonstrate that SR alone can produce alterations to sleep and temperature control that could have consequences for astronauts and their ability to meet mission demands.
Collapse
Affiliation(s)
- Larry D Sanford
- Sleep Research Laboratory, Center for Integrative Neuroscience and Inflammatory Diseases, Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Austin M Adkins
- Sleep Research Laboratory, Center for Integrative Neuroscience and Inflammatory Diseases, Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Alea F Boden
- Sleep Research Laboratory, Center for Integrative Neuroscience and Inflammatory Diseases, Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Justin D Gotthold
- Sleep Research Laboratory, Center for Integrative Neuroscience and Inflammatory Diseases, Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Ryan D Harris
- Sleep Research Laboratory, Center for Integrative Neuroscience and Inflammatory Diseases, Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Dorela Shuboni-Mulligan
- Sleep Research Laboratory, Center for Integrative Neuroscience and Inflammatory Diseases, Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Laurie L Wellman
- Sleep Research Laboratory, Center for Integrative Neuroscience and Inflammatory Diseases, Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Richard A Britten
- Center for Integrative Neuroscience and Inflammatory Diseases, Radiation Oncology, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| |
Collapse
|
11
|
The Effects of Galactic Cosmic Rays on the Central Nervous System: From Negative to Unexpectedly Positive Effects That Astronauts May Encounter. BIOLOGY 2023; 12:biology12030400. [PMID: 36979092 PMCID: PMC10044754 DOI: 10.3390/biology12030400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/23/2023] [Accepted: 02/25/2023] [Indexed: 03/06/2023]
Abstract
Galactic cosmic rays (GCR) pose a serious threat to astronauts’ health during deep space missions. The possible functional alterations of the central nervous system (CNS) under GCR exposure can be critical for mission success. Despite the obvious negative effects of ionizing radiation, a number of neutral or even positive effects of GCR irradiation on CNS functions were revealed in ground-based experiments with rodents and primates. This review is focused on the GCR exposure effects on emotional state and cognition, emphasizing positive effects and their potential mechanisms. We integrate these data with GCR effects on adult neurogenesis and pathological protein aggregation, forming a complete picture. We conclude that GCR exposure causes multidirectional effects on cognition, which may be associated with emotional state alterations. However, the irradiation in space-related doses either has no effect or has performance enhancing effects in solving high-level cognition tasks and tasks with a high level of motivation. We suppose the model of neurotransmission changes after irradiation, although the molecular mechanisms of this phenomenon are not fully understood.
Collapse
|
12
|
Alaghband Y, Klein PM, Kramár EA, Cranston MN, Perry BC, Shelerud LM, Kane AE, Doan NL, Ru N, Acharya MM, Wood MA, Sinclair DA, Dickstein DL, Soltesz I, Limoli CL, Baulch JE. Galactic cosmic radiation exposure causes multifaceted neurocognitive impairments. Cell Mol Life Sci 2023; 80:29. [PMID: 36607431 PMCID: PMC9823026 DOI: 10.1007/s00018-022-04666-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 11/01/2022] [Accepted: 12/11/2022] [Indexed: 01/07/2023]
Abstract
Technological advancements have facilitated the implementation of realistic, terrestrial-based complex 33-beam galactic cosmic radiation simulations (GCR Sim) to now probe central nervous system functionality. This work expands considerably on prior, simplified GCR simulations, yielding new insights into responses of male and female mice exposed to 40-50 cGy acute or chronic radiations relevant to deep space travel. Results of the object in updated location task suggested that exposure to acute or chronic GCR Sim induced persistent impairments in hippocampus-dependent memory formation and reconsolidation in female mice that did not manifest robustly in irradiated male mice. Interestingly, irradiated male mice, but not females, were impaired in novel object recognition and chronically irradiated males exhibited increased aggressive behavior on the tube dominance test. Electrophysiology studies used to evaluate synaptic plasticity in the hippocampal CA1 region revealed significant reductions in long-term potentiation after each irradiation paradigm in both sexes. Interestingly, network-level disruptions did not translate to altered intrinsic electrophysiological properties of CA1 pyramidal cells, whereas acute exposures caused modest drops in excitatory synaptic signaling in males. Ultrastructural analyses of CA1 synapses found smaller postsynaptic densities in larger spines of chronically exposed mice compared to controls and acutely exposed mice. Myelination was also affected by GCR Sim with acutely exposed mice exhibiting an increase in the percent of myelinated axons; however, the myelin sheathes on small calibur (< 0.3 mm) and larger (> 0.5 mm) axons were thinner when compared to controls. Present findings might have been predicted based on previous studies using single and mixed beam exposures and provide further evidence that space-relevant radiation exposures disrupt critical cognitive processes and underlying neuronal network-level plasticity, albeit not to the extent that might have been previously predicted.
Collapse
Affiliation(s)
- Yasaman Alaghband
- Department of Radiation Oncology, Medical Sciences I, University of California Irvine, Room B-146D, Irvine, CA, 92697-2695, USA
| | - Peter M Klein
- Department of Neurosurgery, Stanford University, Palo Alto, CA, 94305, USA
| | - Eniko A Kramár
- Department of Neurobiology and Behavior, School of Biological Sciences, University of California, Irvine, 92697-2695, USA
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, 92697-2695, USA
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, 92697-2695, USA
| | - Michael N Cranston
- Department of Pathology, Uniformed Services University of Health Sciences, Bethesda, MD, 20814, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine (HJF), Bethesda, MD, 20817, USA
| | - Bayley C Perry
- Department of Pathology, Uniformed Services University of Health Sciences, Bethesda, MD, 20814, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine (HJF), Bethesda, MD, 20817, USA
| | - Lukas M Shelerud
- Department of Genetics, Blavatnik Institute, Paul F. Glenn Center for Biology of Aging Research, Harvard Medical School, Boston, MA, 0211, USA
| | - Alice E Kane
- Department of Genetics, Blavatnik Institute, Paul F. Glenn Center for Biology of Aging Research, Harvard Medical School, Boston, MA, 0211, USA
| | - Ngoc-Lien Doan
- Department of Radiation Oncology, Medical Sciences I, University of California Irvine, Room B-146D, Irvine, CA, 92697-2695, USA
| | - Ning Ru
- Department of Radiation Oncology, Medical Sciences I, University of California Irvine, Room B-146D, Irvine, CA, 92697-2695, USA
| | - Munjal M Acharya
- Department of Radiation Oncology, Medical Sciences I, University of California Irvine, Room B-146D, Irvine, CA, 92697-2695, USA
- Department of Anatomy and Neurobiology, University of California, Irvine, 92697-2695, USA
| | - Marcelo A Wood
- Department of Neurobiology and Behavior, School of Biological Sciences, University of California, Irvine, 92697-2695, USA
| | - David A Sinclair
- Department of Genetics, Blavatnik Institute, Paul F. Glenn Center for Biology of Aging Research, Harvard Medical School, Boston, MA, 0211, USA
| | - Dara L Dickstein
- Department of Pathology, Uniformed Services University of Health Sciences, Bethesda, MD, 20814, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine (HJF), Bethesda, MD, 20817, USA
| | - Ivan Soltesz
- Department of Neurosurgery, Stanford University, Palo Alto, CA, 94305, USA
- Department of Neurology and Neurological Sciences, Stanford University, Palo Alto, CA, 94305, USA
| | - Charles L Limoli
- Department of Radiation Oncology, Medical Sciences I, University of California Irvine, Room B-146D, Irvine, CA, 92697-2695, USA
| | - Janet E Baulch
- Department of Radiation Oncology, Medical Sciences I, University of California Irvine, Room B-146D, Irvine, CA, 92697-2695, USA.
| |
Collapse
|
13
|
Simmons P, Trujillo M, McElroy T, Binz R, Pathak R, Allen AR. Evaluating the effects of low-dose simulated galactic cosmic rays on murine hippocampal-dependent cognitive performance. Front Neurosci 2022; 16:908632. [PMID: 36561122 PMCID: PMC9765097 DOI: 10.3389/fnins.2022.908632] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 09/12/2022] [Indexed: 12/12/2022] Open
Abstract
Space exploration has advanced substantially over recent decades and plans to increase the duration of deep space missions are in preparation. One of the primary health concerns is potential damage to the central nervous system (CNS), resulting in loss of cognitive abilities and function. The majority of ground-based research on space radiation-induced health risks has been conducted using single particle simulations, which do not effectively model real-world scenarios. Thus, to improve the safety of space missions, we must expand our understanding of the effects of simulated galactic cosmic rays (GCRs) on the CNS. To assess the effects of low-dose GCR, we subjected 6-month-old male BALB/c mice to 50 cGy 5-beam simplified GCR spectrum (1H, 28Si, 4He, 16O, and 56Fe) whole-body irradiation at the NASA Space Radiation Laboratory. Animals were tested for cognitive performance with Y-maze and Morris water maze tests 3 months after irradiation. Irradiated animals had impaired short-term memory and lacked spatial memory retention on day 5 of the probe trial. Glial cell analysis by flow cytometry showed no significant changes in oligodendrocytes, astrocytes, microglia or neural precursor cells (NPC's) between the sham group and GCR group. Bone marrow cytogenetic data showed a significant increase in the frequency of chromosomal aberrations after GCR exposure. Finally, tandem mass tag proteomics identified 3,639 proteins, 113 of which were differentially expressed when comparing sham versus GCR exposure (fold change > 1.5; p < 0.05). Our data suggest exposure to low-dose GCR induces cognitive deficits by impairing short-term memory and spatial memory retention.
Collapse
Affiliation(s)
- Pilar Simmons
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States,Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Madison Trujillo
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States,Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Taylor McElroy
- Department of Aging, University of Florida, Gainesville, FL, United States
| | - Regina Binz
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Rupak Pathak
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Antiño R. Allen
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States,Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States,*Correspondence: Antiño R. Allen,
| |
Collapse
|
14
|
Rubinstein L, Kiffer F, Puukila S, Lowe MG, Goo B, Luthens A, Schreurs AS, Torres SM, Steczina S, Tahimic CGT, Allen AR. Mitochondria-Targeted Human Catalase in the Mouse Longevity MCAT Model Mitigates Head-Tilt Bedrest-Induced Neuro-Inflammation in the Hippocampus. Life (Basel) 2022; 12:1838. [PMID: 36362993 PMCID: PMC9695374 DOI: 10.3390/life12111838] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/28/2022] [Accepted: 11/03/2022] [Indexed: 10/22/2024] Open
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.
Collapse
Affiliation(s)
- Linda Rubinstein
- Universities Space Research Association USRA, Columbia, MD 21046, USA
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, Mountain View, CA 94035, USA
- The Joseph Sagol Neuroscience Center, Sheba Research Hospital, Ramat Gan 52621, Israel
| | - Frederico Kiffer
- Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Radiation Health Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Stephanie Puukila
- Universities Space Research Association USRA, Columbia, MD 21046, USA
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, Mountain View, CA 94035, USA
| | - Moniece G Lowe
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, Mountain View, CA 94035, USA
- Blue Marble Space Institute of Science, Seattle, WA 98154, USA
| | - Brie Goo
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, Mountain View, CA 94035, USA
| | | | - Ann-Sofie Schreurs
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, Mountain View, CA 94035, USA
| | - Samantha M Torres
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, Mountain View, CA 94035, USA
- Blue Marble Space Institute of Science, Seattle, WA 98154, USA
| | - Sonette Steczina
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, Mountain View, CA 94035, USA
- Blue Marble Space Institute of Science, Seattle, WA 98154, USA
| | - Candice G T Tahimic
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, Mountain View, CA 94035, USA
- KBR, Houston, TX 77002, USA
- Department of Biology, University of North Florida, Jacksonville, FL 32224, USA
| | - Antiño R Allen
- Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Radiation Health Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
15
|
Kokhan VS, Ustyugov AA, Pikalov VA. Dynamics of Dopamine and Other Monoamines Content in Rat Brain after Single Low-Dose Carbon Nuclei Irradiation. Life (Basel) 2022; 12:life12091306. [PMID: 36143343 PMCID: PMC9502711 DOI: 10.3390/life12091306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/10/2022] [Accepted: 08/20/2022] [Indexed: 11/16/2022] Open
Abstract
Space radiation, presented primarily by high-charge and -energy particles (HZEs), has a substantial impact on the central nervous system (CNS) of astronauts. This impact, surprisingly, has not only negative but also positive effects on CNS functions. Despite the fact that the mechanisms of this effect have not yet been elucidated, several studies indicate a key role for monoaminergic networks underlying these effects. Here, we investigated the effects of acute irradiation with 450 MeV/n carbon (12C) nuclei at a dose of 0.14 Gy on Wistar rats; a state of anxiety was accessed using a light–dark box, spatial memory in a Morris water maze, and the dynamics of monoamine metabolism in several brain morphological structures using HPLC. No behavioral changes were observed. Irradiation led to the immediate suppression of dopamine turnover in the prefrontal cortex, hypothalamus, and striatum, while a decrease in the level of norepinephrine was detected in the amygdala. However, these effects were transient. The deferred effect of dopamine turnover increase was found in the hippocampus. These data underscore the ability of even low-dose 12C irradiation to affect monoaminergic networks. However, this impact is transient and is not accompanied by behavioral alterations.
Collapse
Affiliation(s)
- Viktor S. Kokhan
- V.P. Serbsky Federal Medical Research Centre for Psychiatry and Narcology, 119034 Moscow, Russia
- Correspondence: ; Tel.: +7-92-5462-9948
| | - Alexey A. Ustyugov
- Institute of Physiologically Active Compounds RAS, 142432 Chernogolovka, Russia
| | - Vladimir A. Pikalov
- Institute for High Energy Physics Named by A.A. Logunov of National Research Centre “Kurchatov Institute”, 142281 Protvino, Russia
| |
Collapse
|
16
|
Desai RI, Limoli CL, Stark CEL, Stark SM. Impact of spaceflight stressors on behavior and cognition: A molecular, neurochemical, and neurobiological perspective. Neurosci Biobehav Rev 2022; 138:104676. [PMID: 35461987 DOI: 10.1016/j.neubiorev.2022.104676] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 03/15/2022] [Accepted: 04/18/2022] [Indexed: 11/19/2022]
Abstract
The response of the human body to multiple spaceflight stressors is complex, but mounting evidence implicate risks to CNS functionality as significant, able to threaten metrics of mission success and longer-term behavioral and neurocognitive health. Prolonged exposure to microgravity, sleep disruption, social isolation, fluid shifts, and ionizing radiation have been shown to disrupt mechanisms of homeostasis and neurobiological well-being. The overarching goal of this review is to document the existing evidence of how the major spaceflight stressors, including radiation, microgravity, isolation/confinement, and sleep deprivation, alone or in combination alter molecular, neurochemical, neurobiological, and plasma metabolite/lipid signatures that may be linked to operationally-relevant behavioral and cognitive performance. While certain brain region-specific and/or systemic alterations titrated in part with neurobiological outcome, variations across model systems, study design, and the conspicuous absence of targeted studies implementing combinations of spaceflight stressors, confounded the identification of specific signatures having direct relevance to human activities in space. Summaries are provided for formulating new research directives and more predictive readouts of portending change in neurobiological function.
Collapse
Affiliation(s)
- Rajeev I Desai
- Harvard Medical School, McLean Hospital, Behavioral Biology Program, Belmont, MA 02478, USA.
| | - Charles L Limoli
- Department of Radiation Oncology, University of California Irvine, Medical Sciences I, B146B, Irvine, CA 92697, USA
| | - Craig E L Stark
- Department of Neurobiology of Behavior, University of California Irvine, 1400 Biological Sciences III, Irvine, CA 92697, USA
| | - Shauna M Stark
- Department of Neurobiology of Behavior, University of California Irvine, 1400 Biological Sciences III, Irvine, CA 92697, USA
| |
Collapse
|
17
|
Kiffer FC, Luitel K, Tran FH, Patel RA, Guzman CS, Soler I, Xiao R, Shay JW, Yun S, Eisch AJ. Effects of a 33-ion sequential beam galactic cosmic ray analog on male mouse behavior and evaluation of CDDO-EA as a radiation countermeasure. Behav Brain Res 2022; 419:113677. [PMID: 34818568 PMCID: PMC9755463 DOI: 10.1016/j.bbr.2021.113677] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/28/2021] [Accepted: 11/16/2021] [Indexed: 12/21/2022]
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.
Collapse
Affiliation(s)
- Frederico C Kiffer
- Department of Anesthesiology and Critical Care Medicine, The Children’s Hospital of Philadelphia (CHOP) Research Institute, Philadelphia, PA, USA, 19104
| | - Krishna Luitel
- Department of Cell Biology, University of Texas Southwestern (UTSW) Medical Center, Dallas, TX, USA, 75390
| | - Fionya H Tran
- Department of Anesthesiology and Critical Care Medicine, The Children’s Hospital of Philadelphia (CHOP) Research Institute, Philadelphia, PA, USA, 19104
| | - Riya A Patel
- Department of Anesthesiology and Critical Care Medicine, The Children’s Hospital of Philadelphia (CHOP) Research Institute, Philadelphia, PA, USA, 19104
| | - Catalina S Guzman
- Department of Anesthesiology and Critical Care Medicine, The Children’s Hospital of Philadelphia (CHOP) Research Institute, Philadelphia, PA, USA, 19104
| | - Ivan Soler
- Department of Anesthesiology and Critical Care Medicine, The Children’s Hospital of Philadelphia (CHOP) Research Institute, Philadelphia, PA, USA, 19104
| | - Rui Xiao
- Department of Pediatrics Division of Biostatistics, CHOP Research Institute, Philadelphia, PA, USA, 19104,Department of Biostatistics, Epidemiology & Informatics, University of Pennsylvania, Philadelphia, PA, USA, 19104
| | - Jerry W Shay
- Department of Cell Biology, University of Texas Southwestern (UTSW) Medical Center, Dallas, TX, USA, 75390
| | - Sanghee Yun
- Department of Anesthesiology and Critical Care Medicine, The Children’s Hospital of Philadelphia (CHOP) Research Institute, Philadelphia, PA, USA, 19104,Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA, 19104
| | - Amelia J Eisch
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia (CHOP) Research Institute, Philadelphia, PA 19104, USA; Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
18
|
Burket JA, Matar M, Fesshaye A, Pickle JC, Britten RA. Exposure to Low (≤10 cGy) Doses of 4He Particles Leads to Increased Social Withdrawal and Loss of Executive Function Performance. Radiat Res 2021; 196:345-354. [PMID: 34270762 DOI: 10.1667/rade-20-00251.1] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 07/06/2021] [Indexed: 11/03/2022]
Abstract
Astronauts on the planned mission to Mars will be exposed to galactic cosmic radiation (GCR), with proton and He particles accounting (in approximately equal amounts) for ∼75% of the equivalent dose. Exposure to ≤15 cGy of space radiation ions with Z ≥ 15 particles has been shown to impair various executive functions, including attentional set shifting and creative problem-solving in rats. Executive functions also regulate social interactions and mood. Should space radiation exposure alter these executive functions as it does cognitive flexibility, there is the possibility of altered interactions among crew members and team cooperativity during prolonged space exploration. This study characterized the effects of ≤10 cGy 400 MeV/n of 4He particles on cognitive flexibility and social interaction (within freely interacting dyads) in male Wistar rats. Exposure to ≥1 cGy 4He ions induced deficits in the SD and/or CD stages of the attentional set shifting (ATSET) task, as reported after exposure to Z ≥ 15 space radiation ions. Should similar effects occur in astronauts, these data suggest that they would have a reduced ability to identify key events in a new situation and would be more easily distracted by extraneous variables. The irradiated rats were also screened for performance in a task for unconstrained cognitive flexibility (UCFlex), often referred to as creative problem-solving. There was a marked dose-dependent change in UCFlex performance with ∼30% of rats exposed to 10 cGy being unable to solve the problem, while the remaining rats took longer than the sham-irradiated animals to resolve the problem. Importantly, performance in the ATSET test was not indicative of UCFlex performance. From a risk assessment perspective, these findings suggest that a value based on a single behavioral end point may not fully represent the cognitive deficits induced by space radiation, even within the cognitive flexibility domain. Rats that received 5 cGy 4He ion irradiation had a significantly lower level of interaction toward their sham-irradiated partners in a non-anxiogenic (uncaged) dyad interactions study. This is consistent with the social withdrawal previously observed in space radiation-exposed male mice in a three-chamber test. 4He-irradiated rats exhibited a significantly higher incidence and duration of self-grooming, which is even more concerning, given that their dyad partners were able to physically interact with the irradiated rats (i.e., touching/climbing over them). This study has established that exposure of male rats to "light" ions such as He affects multiple executive functions resulting in deficits in both sociability and cognitive flexibility, and possibly affective behavior (reward valuation). Further studies are needed to determine if these space radiation-induced co-morbidities are concomitantly induced within individual rats.
Collapse
Affiliation(s)
- Jessica A Burket
- Department of Molecular Biology and Chemistry, Christopher Newport University, Newport News, Virginia 23606
- Department of Program in Neuroscience, Christopher Newport University, Newport News, Virginia 23606
| | - Mona Matar
- National Aeronautics and Space Administration, John H. Glenn Research Center, Cleveland, Ohio 44135
| | - Arriyam Fesshaye
- Department of Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Jerrah C Pickle
- School of Medicine, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Richard A Britten
- Department of Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, Virginia 23507
- Leroy T Canoles Jr. Cancer Center, Eastern Virginia Medical School, Norfolk, Virginia 23507
- Center for Integrative Neuroinflammatory and Inflammatory diseases, Eastern Virginia Medical School, Norfolk, Virginia 23507
| |
Collapse
|
19
|
Swinton C, Kiffer F, McElroy T, Wang J, Sridharan V, Boerma M, Allen AR. Effects of 16O charged-particle irradiation on cognition, hippocampal morphology and mutagenesis in female mice. Behav Brain Res 2021; 407:113257. [PMID: 33794227 DOI: 10.1016/j.bbr.2021.113257] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 02/23/2021] [Accepted: 03/17/2021] [Indexed: 11/24/2022]
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.
Collapse
Affiliation(s)
- Chase Swinton
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, United States.
| | - Frederico Kiffer
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, United States.
| | - Taylor McElroy
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, United States; Neurobiology & Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, United States.
| | - Jing Wang
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, United States.
| | - Vijayalakshmi Sridharan
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, United States.
| | - Marjan Boerma
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, United States.
| | - Antiño R Allen
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, United States; Neurobiology & Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, United States.
| |
Collapse
|
20
|
Davis CM, Allen AR, Bowles DE. Consequences of space radiation on the brain and cardiovascular system. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART C, TOXICOLOGY AND CARCINOGENESIS 2021; 39:180-218. [PMID: 33902387 DOI: 10.1080/26896583.2021.1891825] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Staying longer in outer space will inevitably increase the health risks of astronauts due to the exposures to galactic cosmic rays and solar particle events. Exposure may pose a significant hazard to space flight crews not only during the mission but also later, when slow-developing adverse effects could finally become apparent. The body of literature examining ground-based outcomes in response to high-energy charged-particle radiation suggests differential effects in response to different particles and energies. Numerous animal and cellular models have repeatedly demonstrated the negative effects of high-energy charged-particle on the brain and cognitive function. However, research on the role of space radiation in potentiating cardiovascular dysfunction is still in its early stages. This review summarizes the available data from studies using ground-based animal models to evaluate the response of the brain and heart to the high-energy charged particles of GCR and SPE, addresses potential sex differences in these effects, and aims to highlight gaps in the current literature for future study.
Collapse
Affiliation(s)
- Catherine M Davis
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences Bethesda, MD, USA
| | - Antiño R Allen
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Dawn E Bowles
- Division of Surgical Sciences, Department of Surgery, Duke University, Durham, NC, USA
| |
Collapse
|
21
|
Parihar VK, Angulo MC, Allen BD, Syage A, Usmani MT, Passerat de la Chapelle E, Amin AN, Flores L, Lin X, Giedzinski E, Limoli CL. Sex-Specific Cognitive Deficits Following Space Radiation Exposure. Front Behav Neurosci 2020; 14:535885. [PMID: 33192361 PMCID: PMC7525092 DOI: 10.3389/fnbeh.2020.535885] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 08/24/2020] [Indexed: 12/17/2022] Open
Abstract
The radiation fields in space define tangible risks to the health of astronauts, and significant work in rodent models has clearly shown a variety of exposure paradigms to compromise central nervous system (CNS) functionality. Despite our current knowledge, sex differences regarding the risks of space radiation exposure on cognitive function remain poorly understood, which is potentially problematic given that 30% of astronauts are women. While work from us and others have demonstrated pronounced cognitive decrements in male mice exposed to charged particle irradiation, here we show that female mice exhibit significant resistance to adverse neurocognitive effects of space radiation. The present findings indicate that male mice exposed to low doses (≤30 cGy) of energetic (400 MeV/n) helium ions (4He) show significantly higher levels of neuroinflammation and more extensive cognitive deficits than females. Twelve weeks following 4He ion exposure, irradiated male mice demonstrated significant deficits in object and place recognition memory accompanied by activation of microglia, marked upregulation of hippocampal Toll-like receptor 4 (TLR4), and increased expression of the pro-inflammatory marker high mobility group box 1 protein (HMGB1). Additionally, we determined that exposure to 4He ions caused a significant decline in the number of dendritic branch points and total dendritic length along with the hippocampus neurons in female mice. Interestingly, only male mice showed a significant decline of dendritic spine density following irradiation. These data indicate that fundamental differences in inflammatory cascades between male and female mice may drive divergent CNS radiation responses that differentially impact the structural plasticity of neurons and neurocognitive outcomes following cosmic radiation exposure.
Collapse
Affiliation(s)
- Vipan K Parihar
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, United States
| | - Maria C Angulo
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, United States
| | - Barrett D Allen
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, United States
| | - Amber Syage
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, United States
| | - Manal T Usmani
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, United States
| | | | - Amal Nayan Amin
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, United States
| | - Lidia Flores
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, United States
| | - Xiaomeng Lin
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, United States
| | - Erich Giedzinski
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, United States
| | - Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
22
|
Newton J, Brown T, Corley C, Alexander T, Trujillo M, McElroy T, Ntagwabira F, Wang J, Byrum SD, Allen AR. Cranial irradiation impairs juvenile social memory and modulates hippocampal physiology. Brain Res 2020; 1748:147095. [PMID: 32896524 DOI: 10.1016/j.brainres.2020.147095] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 07/27/2020] [Accepted: 08/27/2020] [Indexed: 01/20/2023]
Abstract
Cranial and craniospinal irradiation are the oldest central nervous system prophylaxis treatments considered for pediatric patients with acute lymphoblastic leukemia (ALL). However, survivors of childhood ALL that received cranial radiotherapy are at increased risk for deficits in neurocognitive skills. The continuous and dynamic response of normal tissue after irradiation has been identified as one of the causative factors for cognitive changes after cranial radiation therapy. The aim of our study was to investigate the radiation effects on social behavior and neuronal morphology in the hippocampus of adult mice. Twenty-oneday-old male C57BL/6 mice were irradiated with the small-animal radiation research platform (SARRP). Animals were given a single 10-Gy dose of radiation of X-ray cranial radiation. One month following irradiation, animals underwent behavioral testing in the Three-Chamber Sociability paradigm. Radiation affected social discrimination during the third stage eliciting an inability to discriminate between the familiar and stranger mouse, while sham successfully spent more time exploring the novel stranger. Proteomic analysis revealed dysregulation of metabolic and signaling pathways associated with neurocognitive dysfunction such as mitochondrial dysfunction, Rac 1 signaling, and synaptogenesis signaling. We observed significant decreases in mushroom spine density in the Cornu Ammonis 2 of the hippocampus, which is associated with sociability processing.
Collapse
Affiliation(s)
- Jamila Newton
- California State University, Stanislaus, Turlock, CA 95382, United States
| | - Taurean Brown
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Christa Corley
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Tyler Alexander
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Madison Trujillo
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Taylor McElroy
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Fabio Ntagwabira
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Jing Wang
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Stephanie D Byrum
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States; Arkansas Children's Research Institute, Little Rock, AR 72202, United States
| | - Antiño R Allen
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States; Neurobiology & Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States.
| |
Collapse
|
23
|
Kiffer F, Alexander T, Anderson J, Groves T, McElroy T, Wang J, Sridharan V, Bauer M, Boerma M, Allen A. Late Effects of 1H + 16O on Short-Term and Object Memory, Hippocampal Dendritic Morphology and Mutagenesis. Front Behav Neurosci 2020; 14:96. [PMID: 32670032 PMCID: PMC7332779 DOI: 10.3389/fnbeh.2020.00096] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 05/22/2020] [Indexed: 11/17/2022] Open
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.
Collapse
Affiliation(s)
- Frederico Kiffer
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Tyler Alexander
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Julie Anderson
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Thomas Groves
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Neurobiology & Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Taylor McElroy
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Jing Wang
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Vijayalakshmi Sridharan
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Michael Bauer
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Marjan Boerma
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Antiño Allen
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Neurobiology & Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| |
Collapse
|
24
|
McElroy T, Brown T, Kiffer F, Wang J, Byrum SD, Oberley-Deegan RE, Allen AR. Assessing the Effects of Redox Modifier MnTnBuOE-2-PyP 5+ on Cognition and Hippocampal Physiology Following Doxorubicin, Cyclophosphamide, and Paclitaxel Treatment. Int J Mol Sci 2020; 21:ijms21051867. [PMID: 32182883 PMCID: PMC7084440 DOI: 10.3390/ijms21051867] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/06/2020] [Accepted: 03/07/2020] [Indexed: 12/11/2022] Open
Abstract
Background: Chemotherapy treatment for breast cancer can induce cognitive impairments often involving oxidative stress. The brain, as a whole, is susceptible to oxidative stress due to its high-energy requirements, limited anaerobic respiration capacities, and limited antioxidant defenses. The goal of the current study was to determine if the manganese porphyrin superoxide dismutase mimetic MnTnBuOE-2-PyP (MnBuOE) could ameliorate the effects of doxorubicin, cyclophosphamide, and paclitaxel (AC-T) on mature dendrite morphology and cognitive function. Methods: Four-month-old female C57BL/6 mice received intraperitoneal injections of chemotherapy followed by subcutaneous injections of MnBuOE. Four weeks following chemotherapy treatment, mice were tested for hippocampus-dependent cognitive performance in the Morris water maze. After testing, brains were collected for Golgi staining and molecular analyses. Results: MnBuOE treatment preserved spatial memory during the Morris water-maze. MnBuOE/AC-T showed spatial memory retention during all probe trials. AC-T treatment significantly impaired spatial memory retention in the first and third probe trial (no platform). AC-T treatment decreased dendritic length in the Cornu Ammonis 1 (CA1) and dentate gyrus (DG) areas of the hippocampus while AC-T/MnBuOE maintained dendritic length. Comparative proteomic analysis revealed affected protein networks associated with cell morphology and behavior functions in both the AC-T and AC-T/MnBuOE treatment groups.
Collapse
Affiliation(s)
- Taylor McElroy
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (T.M.); (T.B.); (F.K.); (J.W.)
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Neurobiology & Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Taurean Brown
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (T.M.); (T.B.); (F.K.); (J.W.)
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Neurobiology & Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Fred Kiffer
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (T.M.); (T.B.); (F.K.); (J.W.)
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Jing Wang
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (T.M.); (T.B.); (F.K.); (J.W.)
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Stephanie D. Byrum
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
- Arkansas Children’s Research Institute, Little Rock, AR 72202, USA
| | - Rebecca E. Oberley-Deegan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Antiño R. Allen
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (T.M.); (T.B.); (F.K.); (J.W.)
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Neurobiology & Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Correspondence: ; Tel.: +501-686-7553
| |
Collapse
|
25
|
Liu B, Hinshaw RG, Le KX, Park MA, Wang S, Belanger AP, Dubey S, Frost JL, Shi Q, Holton P, Trojanczyk L, Reiser V, Jones PA, Trigg W, Di Carli MF, Lorello P, Caldarone BJ, Williams JP, O'Banion MK, Lemere CA. Space-like 56Fe irradiation manifests mild, early sex-specific behavioral and neuropathological changes in wildtype and Alzheimer's-like transgenic mice. Sci Rep 2019; 9:12118. [PMID: 31431669 PMCID: PMC6702228 DOI: 10.1038/s41598-019-48615-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 08/06/2019] [Indexed: 12/19/2022] Open
Abstract
Space travel will expose people to high-energy, heavy particle radiation, and the cognitive deficits induced by this exposure are not well understood. To investigate the short-term effects of space radiation, we irradiated 4-month-old Alzheimer’s disease (AD)-like transgenic (Tg) mice and wildtype (WT) littermates with a single, whole-body dose of 10 or 50 cGy 56Fe ions (1 GeV/u) at Brookhaven National Laboratory. At ~1.5 months post irradiation, behavioural testing showed sex-, genotype-, and dose-dependent changes in locomotor activity, contextual fear conditioning, grip strength, and motor learning, mainly in Tg but not WT mice. There was little change in general health, depression, or anxiety. Two months post irradiation, microPET imaging of the stable binding of a translocator protein ligand suggested no radiation-specific change in neuroinflammation, although initial uptake was reduced in female mice independently of cerebral blood flow. Biochemical and immunohistochemical analyses revealed that radiation reduced cerebral amyloid-β levels and microglia activation in female Tg mice, modestly increased microhemorrhages in 50 cGy irradiated male WT mice, and did not affect synaptic marker levels compared to sham controls. Taken together, we show specific short-term changes in neuropathology and behaviour induced by 56Fe irradiation, possibly having implications for long-term space travel.
Collapse
Affiliation(s)
- Bin Liu
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, 02115, USA.,Harvard Medical School, Boston, MA, 02115, USA
| | - Robert G Hinshaw
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, 02115, USA.,Harvard Medical School, Boston, MA, 02115, USA.,Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Kevin X Le
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Mi-Ae Park
- Harvard Medical School, Boston, MA, 02115, USA.,Department of Radiology, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Shuyan Wang
- Department of Radiology, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Anthony P Belanger
- Harvard Medical School, Boston, MA, 02115, USA.,Department of Radiology, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Shipra Dubey
- Harvard Medical School, Boston, MA, 02115, USA.,Department of Radiology, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Jeffrey L Frost
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Qiaoqiao Shi
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, 02115, USA.,Harvard Medical School, Boston, MA, 02115, USA
| | - Peter Holton
- Harvard Medical School, Boston, MA, 02115, USA.,Department of Radiology, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Lee Trojanczyk
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | | | - Paul A Jones
- GE Healthcare, Chalfont St Giles, HP8 4SP, United Kingdom
| | - William Trigg
- GE Healthcare, Chalfont St Giles, HP8 4SP, United Kingdom
| | - Marcelo F Di Carli
- Harvard Medical School, Boston, MA, 02115, USA.,Department of Radiology, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Paul Lorello
- Harvard Medical School Mouse Behavior Core, Boston, MA, 02115, USA
| | | | - Jacqueline P Williams
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - M Kerry O'Banion
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Cynthia A Lemere
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, 02115, USA. .,Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
26
|
Kiffer F, Boerma M, Allen A. Behavioral effects of space radiation: A comprehensive review of animal studies. LIFE SCIENCES IN SPACE RESEARCH 2019; 21:1-21. [PMID: 31101151 PMCID: PMC7150604 DOI: 10.1016/j.lssr.2019.02.004] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 02/14/2019] [Accepted: 02/17/2019] [Indexed: 05/04/2023]
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.
Collapse
Affiliation(s)
- Frederico Kiffer
- Division of Radiation Health, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, United States; Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, United States.
| | - Marjan Boerma
- Division of Radiation Health, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, United States; Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, United States.
| | - Antiño Allen
- Division of Radiation Health, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, United States; Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, United States; Department of Neurobiology & Developmental Sciences, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States.
| |
Collapse
|
27
|
Loss of C/EBPδ Exacerbates Radiation-Induced Cognitive Decline in Aged Mice due to Impaired Oxidative Stress Response. Int J Mol Sci 2019; 20:ijms20040885. [PMID: 30781689 PMCID: PMC6412914 DOI: 10.3390/ijms20040885] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/31/2019] [Accepted: 02/11/2019] [Indexed: 12/20/2022] Open
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.
Collapse
|