1
|
Desai RI, Kangas BD, Luc OT, Solakidou E, Smith EC, Dawes MH, Ma X, Makriyannis A, Chatterjee S, Dayeh MA, Muñoz-Jaramillo A, Desai MI, Limoli CL. Complex 33-beam simulated galactic cosmic radiation exposure impacts cognitive function and prefrontal cortex neurotransmitter networks in male mice. Nat Commun 2023; 14:7779. [PMID: 38012180 PMCID: PMC10682413 DOI: 10.1038/s41467-023-42173-x] [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: 02/10/2023] [Accepted: 09/28/2023] [Indexed: 11/29/2023] Open
Abstract
Astronauts will encounter extended exposure to galactic cosmic radiation (GCR) during deep space exploration, which could impair brain function. Here, we report that in male mice, acute or chronic GCR exposure did not modify reward sensitivity but did adversely affect attentional processes and increased reaction times. Potassium (K+)-stimulation in the prefrontal cortex (PFC) elevated dopamine (DA) but abolished temporal DA responsiveness after acute and chronic GCR exposure. Unlike acute GCR, chronic GCR increased levels of all other neurotransmitters, with differences evident between groups after higher K+-stimulation. Correlational and machine learning analysis showed that acute and chronic GCR exposure differentially reorganized the connection strength and causation of DA and other PFC neurotransmitter networks compared to controls which may explain space radiation-induced neurocognitive deficits.
Collapse
Affiliation(s)
- Rajeev I Desai
- Department of Psychiatry, Harvard Medical School, Boston, MA, 02115, USA.
- Behavioral Biology Program, McLean Hospital, Belmont, MA, 02478, USA.
- Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, 02115, USA.
| | - Brian D Kangas
- Department of Psychiatry, Harvard Medical School, Boston, MA, 02115, USA
- Behavioral Biology Program, McLean Hospital, Belmont, MA, 02478, USA
| | - Oanh T Luc
- Department of Psychiatry, Harvard Medical School, Boston, MA, 02115, USA
- Behavioral Biology Program, McLean Hospital, Belmont, MA, 02478, USA
| | - Eleana Solakidou
- Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, 02115, USA
- Medical School, University of Crete, Heraklion, Greece
| | - Evan C Smith
- Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, 02115, USA
| | - Monica H Dawes
- Department of Psychiatry, Harvard Medical School, Boston, MA, 02115, USA
- Behavioral Biology Program, McLean Hospital, Belmont, MA, 02478, USA
| | - Xiaoyu Ma
- Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, 02115, USA
| | - Alexandros Makriyannis
- Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, 02115, USA
| | | | - Maher A Dayeh
- Southwest Research Institute, San Antonio, TX, 78238, USA
- University of San Antonio, San Antonio, TX, 78249, USA
| | | | - Mihir I Desai
- Southwest Research Institute, San Antonio, TX, 78238, USA
- University of San Antonio, San Antonio, TX, 78249, USA
| | - Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine, Orange, CA, 92697, USA
| |
Collapse
|
2
|
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
|
3
|
Britten RA, Limoli CL. New Radiobiological Principles for the CNS Arising from Space Radiation Research. Life (Basel) 2023; 13:1293. [PMID: 37374076 DOI: 10.3390/life13061293] [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: 03/27/2023] [Revised: 05/17/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
Traditionally, the brain has been regarded as a relatively insensitive late-reacting tissue, with radiologically detectable damage not being reported at doses < 60 Gy. When NASA proposed interplanetary exploration missions, it was required to conduct an intensive health and safety evaluation of cancer, cardiovascular, and cognitive risks associated with exposure to deep space radiation (SR). The SR dose that astronauts on a mission to Mars are predicted to receive is ~300 mGy. Even after correcting for the higher RBE of the SR particles, the biologically effective SR dose (<1 Gy) would still be 60-fold lower than the threshold dose for clinically detectable neurological damage. Unexpectedly, the NASA-funded research program has consistently reported that low (<250 mGy) doses of SR induce deficits in multiple cognitive functions. This review will discuss these findings and the radical paradigm shifts in radiobiological principles for the brain that were required in light of these findings. These included a shift from cell killing to loss of function models, an expansion of the critical brain regions for radiation-induced cognitive impediments, and the concept that the neuron may not be the sole critical target for neurocognitive impairment. The accrued information on how SR exposure impacts neurocognitive performance may provide new opportunities to reduce neurocognitive impairment in brain cancer patients.
Collapse
Affiliation(s)
- Richard A Britten
- EVMS Radiation Oncology, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Charles L Limoli
- Department Radiation Oncology, University of California-Irvine, Irvine, CA 92697, USA
| |
Collapse
|
4
|
Stephenson S, Liu A, Blackwell AA, Britten RA. Multiple decrements in switch task performance in female rats exposed to space radiation. Behav Brain Res 2023; 449:114465. [PMID: 37142163 DOI: 10.1016/j.bbr.2023.114465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/14/2023] [Accepted: 05/01/2023] [Indexed: 05/06/2023]
Abstract
Astronauts on the Artemis missions to the Moon and Mars will be exposed to unavoidable Galactic Cosmic Radiation (GCR). Studies using male rats suggest that GCR exposure impairs several processes required for cognitive flexibility performance, including attention and task switching. Currently no comparable studies have been conducted with female rats. Given that both males and females will travel into deep space, this study determined whether simulated GCR (GCRsim) exposure impairs task switching performance in female rats. Female Wistar rats exposed to 10cGy GCRsim (n = 12) and shams (n=14) were trained to perform a touchscreen-based switch task that mimics a switch task used to evaluate pilots' response times. In comparison to sham rats, three-fold more GCRsim-exposed rats failed to complete the stimulus response stage of training, a high cognitive loading task. In the switch task, 50% of the GCRsim-exposed rats failed to consistently transition between the repeated and switch blocks of stimuli, which they completed during lower cognitive loading training stages. The GCRsim-exposed rats that completed the switch task only performed at 65% of the accuracy of shams. Female rats exposed to GCRsim thus exhibit multiple decrements in the switch task under high, but not low, cognitive loading conditions. While the operational significance of this performance decrement is unknown, if GCRSim exposure was to induce similar effects in astronauts, our data suggests there may be a reduced ability to execute task switching under high cognitive loading situations.
Collapse
Affiliation(s)
- Samuel Stephenson
- School of Medicine, Eastern Virginia Medical School, Norfolk, Virginia 23507 USA
| | - Aiyi Liu
- EVMS Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507 USA
| | - Ashley A Blackwell
- EVMS Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507 USA; Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, Virginia 23507 USA
| | - Richard A Britten
- EVMS Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507 USA; Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, Virginia 23507 USA.
| |
Collapse
|
5
|
Hinshaw RG, Schroeder MK, Ciola J, Varma C, Colletti B, Liu B, Liu GG, Shi Q, Williams JP, O’Banion MK, Caldarone BJ, Lemere CA. High-Energy, Whole-Body Proton Irradiation Differentially Alters Long-Term Brain Pathology and Behavior Dependent on Sex and Alzheimer's Disease Mutations. Int J Mol Sci 2023; 24:ijms24043615. [PMID: 36835027 PMCID: PMC9965515 DOI: 10.3390/ijms24043615] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 01/28/2023] [Accepted: 01/31/2023] [Indexed: 02/17/2023] Open
Abstract
Whole-body exposure to high-energy particle radiation remains an unmitigated hazard to human health in space. Ongoing experiments at the NASA Space Radiation Laboratory and elsewhere repeatedly show persistent changes in brain function long after exposure to simulations of this unique radiation environment, although, as is also the case with proton radiotherapy sequelae, how this occurs and especially how it interacts with common comorbidities is not well-understood. Here, we report modest differential changes in behavior and brain pathology between male and female Alzheimer's-like and wildtype littermate mice 7-8 months after exposure to 0, 0.5, or 2 Gy of 1 GeV proton radiation. The mice were examined with a battery of behavior tests and assayed for amyloid beta pathology, synaptic markers, microbleeds, microglial reactivity, and plasma cytokines. In general, the Alzheimer's model mice were more prone than their wildtype littermates to radiation-induced behavior changes, and hippocampal staining for amyloid beta pathology and microglial activation in these mice revealed a dose-dependent reduction in males but not in females. In summary, radiation-induced, long-term changes in behavior and pathology, although modest, appear specific to both sex and the underlying disease state.
Collapse
Affiliation(s)
- Robert G. Hinshaw
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02129, USA
| | - Maren K. Schroeder
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Jason Ciola
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Curran Varma
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Brianna Colletti
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Bin Liu
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Departments of Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Grace Geyu Liu
- 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
- Departments of Neurology, Harvard Medical School, 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, Del Monte Institute 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
- Departments of Neurology, Harvard Medical School, Boston, MA 02115, USA
- Correspondence:
| |
Collapse
|
6
|
Comparative Analysis of Behavioral Reactions and Morphological Changes in the Rat Brain After Exposure to Ionizing Radiation with Different Physical Characteristics. Cell Mol Neurobiol 2023; 43:339-353. [PMID: 34982311 DOI: 10.1007/s10571-021-01187-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 12/24/2021] [Indexed: 01/11/2023]
Abstract
The aim of this research was to study behavioral reactions and morphological changes in the brain of adult female Sprague Dawley rats after exposure to 170 MeV and 70 MeV protons and gamma radiation (60Co) at a dose of 1 Gy. The analysis of the behavioral reactions in the T-maze showed that exposure to ionizing radiation with different LETs led to an increase in number of repeated entries into the arms of the maze in the spontaneous alternation test. In the Open Field test a decrease in overall motor activity in the group of irradiated animals (70 MeV protons at the Bragg peak) was observed. A decrease in the number of standing positions was seen in all groups of irradiated animals. Morphological analysis showed the development of early amyloidosis, autolysis of the ependymal layer, an increase in the number of neurodegenerative changes in various structures of the brain, and the development of neuronal hypertrophy on the 30th day after irradiation in the cerebellum and hippocampal hilus. Exposure to protons at a dose of 1 Gy leads to the development of structural and functional disorders of the central nervous system of animals on the 30th day after irradiation. These data indicate a damage of short-term memory, a decrease in motor activity and exploratory behavior of animals. With an increase in LET, there is an increase in the number of amyloid plaques in the forebrain of rats, autolysis of the ependymal layer of the ventricles, and the development of dystrophic changes. Investigations of behavioral reactions and morphological changes in various parts of the brain of adult rats on the 30th day after influence of ionizing radiation with different physical characteristics at a dose of 1 Gy. Various negative patho-morphological and cognitive-behavioral changes observed.
Collapse
|
7
|
Miller KB, Mi KL, Nelson GA, Norman RB, Patel ZS, Huff JL. Ionizing radiation, cerebrovascular disease, and consequent dementia: A review and proposed framework relevant to space radiation exposure. Front Physiol 2022; 13:1008640. [PMID: 36388106 PMCID: PMC9640983 DOI: 10.3389/fphys.2022.1008640] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 09/29/2022] [Indexed: 09/05/2023] Open
Abstract
Space exploration requires the characterization and management or mitigation of a variety of human health risks. Exposure to space radiation is one of the main health concerns because it has the potential to increase the risk of cancer, cardiovascular disease, and both acute and late neurodegeneration. Space radiation-induced decrements to the vascular system may impact the risk for cerebrovascular disease and consequent dementia. These risks may be independent or synergistic with direct damage to central nervous system tissues. The purpose of this work is to review epidemiological and experimental data regarding the impact of low-to-moderate dose ionizing radiation on the central nervous system and the cerebrovascular system. A proposed framework outlines how space radiation-induced effects on the vasculature may increase risk for both cerebrovascular dysfunction and neural and cognitive adverse outcomes. The results of this work suggest that there are multiple processes by which ionizing radiation exposure may impact cerebrovascular function including increases in oxidative stress, neuroinflammation, endothelial cell dysfunction, arterial stiffening, atherosclerosis, and cerebral amyloid angiopathy. Cerebrovascular adverse outcomes may also promote neural and cognitive adverse outcomes. However, there are many gaps in both the human and preclinical evidence base regarding the long-term impact of ionizing radiation exposure on brain health due to heterogeneity in both exposures and outcomes. The unique composition of the space radiation environment makes the translation of the evidence base from terrestrial exposures to space exposures difficult. Additional investigation and understanding of the impact of low-to-moderate doses of ionizing radiation including high (H) atomic number (Z) and energy (E) (HZE) ions on the cerebrovascular system is needed. Furthermore, investigation of how decrements in vascular systems may contribute to development of neurodegenerative diseases in independent or synergistic pathways is important for protecting the long-term health of astronauts.
Collapse
Affiliation(s)
| | | | - Gregory A. Nelson
- Department of Basic Sciences, Division of Biomedical Engineering Sciences, Loma Linda University, Loma Linda, CA, United States
- NASA Johnson Space Center, Houston, TX, United States
- KBR Inc., Houston, TX, United States
| | - Ryan B. Norman
- NASA Langley Research Center, Hampton, VA, United States
| | - Zarana S. Patel
- NASA Johnson Space Center, Houston, TX, United States
- KBR Inc., Houston, TX, United States
| | - Janice L. Huff
- NASA Langley Research Center, Hampton, VA, United States
| |
Collapse
|
8
|
Laiakis EC, Pinheiro M, Nguyen T, Nguyen H, Beheshti A, Dutta SM, Russell WK, Emmett MR, Britten RA. Quantitative proteomic analytic approaches to identify metabolic changes in the medial prefrontal cortex of rats exposed to space radiation. Front Physiol 2022; 13:971282. [PMID: 36091373 PMCID: PMC9459391 DOI: 10.3389/fphys.2022.971282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 07/20/2022] [Indexed: 11/13/2022] Open
Abstract
NASA’s planned mission to Mars will result in astronauts being exposed to ∼350 mSv/yr of Galactic Cosmic Radiation (GCR). A growing body of data from ground-based experiments indicates that exposure to space radiation doses (approximating those that astronauts will be exposed to on a mission to Mars) impairs a variety of cognitive processes, including cognitive flexibility tasks. Some studies report that 33% of individuals may experience severe cognitive impairment. Translating the results from ground-based rodent studies into tangible risk estimates for astronauts is an enormous challenge, but it would be germane for NASA to use the vast body of data from the rodent studies to start developing appropriate countermeasures, in the expectation that some level of space radiation (SR) -induced cognitive impairment could occur in astronauts. While some targeted studies have reported radiation-induced changes in the neurotransmission properties and/or increased neuroinflammation within space radiation exposed brains, there remains little information that can be used to start the development of a mechanism-based countermeasure strategy. In this study, we have employed a robust label-free mass spectrometry (MS) -based untargeted quantitative proteomic profiling approach to characterize the composition of the medial prefrontal cortex (mPFC) proteome in rats that have been exposed to 15 cGy of 600 MeV/n28Si ions. A variety of analytical techniques were used to mine the generated expression data, which in such studies is typically hampered by low and variable sample size. We have identified several pathways and proteins whose expression alters as a result of space radiation exposure, including decreased mitochondrial function, and a further subset of proteins differs in rats that have a high level of cognitive performance after SR exposure in comparison with those that have low performance levels. While this study has provided further insight into how SR impacts upon neurophysiology, and what adaptive responses can be invoked to prevent the emergence of SR-induced cognitive impairment, the main objective of this paper is to outline strategies that can be used by others to analyze sub-optimal data sets and to identify new information.
Collapse
Affiliation(s)
- Evagelia C. Laiakis
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, United States
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC, United States
- *Correspondence: Evagelia C. Laiakis,
| | - Maisa Pinheiro
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, United States
| | - Tin Nguyen
- Department of Computer Science and Engineering, University of Nevada, Reno, NV, United States
| | - Hung Nguyen
- Department of Computer Science and Engineering, University of Nevada, Reno, NV, United States
| | - Afshin Beheshti
- KBR, Space Biosciences Division, NASA Ames Research Center, Moffett Field, Mountain View, CA, United States
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Sucharita M. Dutta
- Department of Obstetrics and Gynecology, Eastern Virginia Medical School, Norfolk, VA, United States
| | - William K. Russell
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, United States
| | - Mark R. Emmett
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, United States
- Department of Radiation Oncology, University of Texas Medical Branch, Galveston, TX, United States
| | - Richard A. Britten
- Department of Radiation Oncology, Eastern Virginia Medical School, Norfolk, VA, United States
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, United States
- Center for Integrative Neuroinflammatory and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA, United States
| |
Collapse
|
9
|
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: 8.0] [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
|
10
|
Ton ST, Laghi JR, Tsai SY, Blackwell AA, Adamczyk NS, Oltmanns JRO, Britten RA, Wallace DG, Kartje GL. Exposure to 5 cGy 28Si Particles Induces Long-Term Microglial Activation in the Striatum and Subventricular Zone and Concomitant Neurogenic Suppression. Radiat Res 2022; 198:28-39. [DOI: 10.1667/rade-21-00021.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 03/17/2022] [Indexed: 11/03/2022]
Abstract
The proposed mission to Mars will expose astronauts to space radiation that is known to adversely affect cognition and tasks that rely on fine sensorimotor function. Space radiation has also been shown to affect the microglial and neurogenic responses in the center nervous system (CNS). We recently reported that a low dose of 5 cGy 600 MeV/n 28Si results in impaired cognition and skilled motor behavior in adult rats. Since these tasks rely at least in part on the proper functioning of the striatum, we examined striatal microglial cells in these same subjects. Using morphometric analysis, we found that 28Si exposure increased activated microglial cells in the striatum. The majority of these striatal Iba1+ microglia were ED1–, indicating that they were in an alternatively activated state, where microglia do not have phagocytic activity but may be releasing cytokines that could negatively impact neuronal function. In the other areas studied, Iba1+ microglial cells were increased in the subventricular zone (SVZ), but not in the dentate gyrus (DG). Additionally, we examined the relationship between the microglial response and neurogenesis. An analysis of new neurons in the DG revealed an increase in doublecortin-positive (DCX+) hilar ectopic granule cells (hEGC) which correlated with Iba1+ cells, suggesting that microglial cells contributed to this aberrant distribution which may adversely affect hippocampal function. Taken together, these results indicate that a single dose of 28Si radiation results in persistent cellular effects in the CNS that may impact astronauts both in the short and long-term following deep space missions.
Collapse
Affiliation(s)
- Son T. Ton
- Research Service, Edward Hines Jr. VA Hospital, Hines, Illinois
| | - Julia R. Laghi
- Research Service, Edward Hines Jr. VA Hospital, Hines, Illinois
| | - Shih-Yen Tsai
- Research Service, Edward Hines Jr. VA Hospital, Hines, Illinois
| | | | | | | | - Richard A. Britten
- Department of Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia
| | - Douglas G. Wallace
- Department of Psychology, Northern Illinois University, DeKalb, Illinois
| | - Gwendolyn L. Kartje
- Research Service, Edward Hines Jr. VA Hospital, Hines, Illinois
- Department of Molecular Pharmacology and Neuroscience, Loyola University Chicago Health Sciences Division, Maywood, Illinois
| |
Collapse
|
11
|
Long-Term Sex- and Genotype-Specific Effects of 56Fe Irradiation on Wild-Type and APPswe/PS1dE9 Transgenic Mice. Int J Mol Sci 2021; 22:ijms222413305. [PMID: 34948098 PMCID: PMC8703695 DOI: 10.3390/ijms222413305] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/07/2021] [Accepted: 12/07/2021] [Indexed: 12/22/2022] Open
Abstract
Space radiation presents a substantial threat to travel beyond Earth. Relatively low doses of high-energy particle radiation cause physiological and behavioral impairments in rodents and may pose risks to human spaceflight. There is evidence that 56Fe irradiation, a significant component of space radiation, may be more harmful to males than to females and worsen Alzheimer's disease pathology in genetically vulnerable models. Yet, research on the long-term, sex- and genotype-specific effects of 56Fe irradiation is lacking. Here, we irradiated 4-month-old male and female, wild-type and Alzheimer's-like APP/PS1 mice with 0, 0.10, or 0.50 Gy of 56Fe ions (1GeV/u). Mice underwent microPET scans before and 7.5 months after irradiation, a battery of behavioral tests at 11 months of age and were sacrificed for pathological and biochemical analyses at 12 months of age. 56Fe irradiation worsened amyloid-beta (Aβ) pathology, gliosis, neuroinflammation and spatial memory, but improved motor coordination, in male transgenic mice and worsened fear memory in wild-type males. Although sham-irradiated female APP/PS1 mice had more cerebral Aβ and gliosis than sham-irradiated male transgenics, female mice of both genotypes were relatively spared from radiation effects 8 months later. These results provide evidence for sex-specific, long-term CNS effects of space radiation.
Collapse
|
12
|
Desai RI, Kangas BD, Limoli CL. Nonhuman primate models in the study of spaceflight stressors: Past contributions and future directions. LIFE SCIENCES IN SPACE RESEARCH 2021; 30:9-23. [PMID: 34281669 DOI: 10.1016/j.lssr.2021.03.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/28/2021] [Accepted: 03/31/2021] [Indexed: 06/13/2023]
Abstract
Studies in rodents suggest that exposure to distinct spaceflight stressors (e.g., space radiation, isolation/confinement, microgravity) may have a profound impact on an astronaut's ability to perform both simple and complex tasks related to neurocognitive performance, central nervous system (CNS) and vestibular/sensorimotor function. However, limited information is currently available on how combined exposure to the spaceflight stressors will impact CNS-related neurocognitive and neurobiological function in-flight and, as well, terrestrial risk of manifesting neurodegenerative conditions when astronauts return to earth. This information gap has significantly hindered our ability to realistically estimate spaceflight hazard risk to the CNS associated with deep space exploration. Notwithstanding a significant body of work with rodents, there have been very few direct investigations of the impact of these spaceflight stressors in combination and, to our knowledge, no such investigations using nonhuman primate (NHP) animal models. In view of the widely-recognized translational value of NHP data in advancing biomedical discoveries, this research deficiency limits our understanding regarding the impact of individual and combined spaceflight stressors on CNS-related neurobiological function. In this review, we address this knowledge gap by conducting a systematic and comprehensive evaluation of existing research on the impact of exposure to spaceflight stressors on NHP CNS-related function. This review is structured to: a) provide an overarching view of the past contributions of NHPs to spaceflight research as well as the strengths, limitations, and translational value of NHP research in its own right and within the existing context of NASA-relevant rodent research; b) highlight specific conclusions based on the published literature and areas needed for future endeavors; c) describe critical research gaps and priorities in NHP research to facilitate NASA's efforts to bridge the key knowledge gaps that currently exist in translating rodent data to humans; and d) provide a roadmap of recommendations for NASA regarding the availability, validity, strengths, and limitations of various NHP models for future targeted research.
Collapse
Affiliation(s)
- Rajeev I Desai
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA.
| | - Brian D Kangas
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine, CA, USA
| |
Collapse
|
13
|
Chronic Low Dose Neutron Exposure Results in Altered Neurotransmission Properties of the Hippocampus-Prefrontal Cortex Axis in Both Mice and Rats. Int J Mol Sci 2021; 22:ijms22073668. [PMID: 33915974 PMCID: PMC8036585 DOI: 10.3390/ijms22073668] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 12/18/2022] Open
Abstract
The proposed deep space exploration to the moon and later to Mars will result in astronauts receiving significant chronic exposures to space radiation (SR). SR exposure results in multiple neurocognitive impairments. Recently, our cross-species (mouse/rat) studies reported impaired associative memory formation in both species following a chronic 6-month low dose exposure to a mixed field of neutrons (1 mGy/day for a total dose pf 18 cGy). In the present study, we report neutron exposure induced synaptic plasticity in the medial prefrontal cortex, accompanied by microglial activation and significant synaptic loss in the hippocampus. In a parallel study, neutron exposure was also found to alter fluorescence assisted single synaptosome LTP (FASS-LTP) in the hippocampus of rats, that may be related to a reduced ability to insert AMPAR into the post-synaptic membrane, which may arise from increased phosphorylation of the serine 845 residue of the GluA1 subunit. Thus, we demonstrate for the first time, that low dose chronic neutron irradiation impacts homeostatic synaptic plasticity in the hippocampal-cortical circuit in two rodent species, and that the ability to successfully encode associative recognition memory is a dynamic, multicircuit process, possibly involving compensatory changes in AMPAR density on the synaptic surface.
Collapse
|
14
|
Britten RA, Wellman LL, Sanford LD. Progressive increase in the complexity and translatability of rodent testing to assess space-radiation induced cognitive impairment. Neurosci Biobehav Rev 2021; 126:159-174. [PMID: 33766676 DOI: 10.1016/j.neubiorev.2021.01.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 12/15/2020] [Accepted: 01/07/2021] [Indexed: 11/29/2022]
Abstract
Ground-based rodent models have established that space radiation doses (approximately those that astronauts will be exposed to on a mission to Mars) significantly impair performance in a wide range of cognitive tasks. Over the last 40 years there has been a progressive increase in both the complexity and the translatability (to humans) of the cognitive tasks investigated. This review outlines technical and conceptual advances in space radiation rodent testing approaches, along with the advances in analytical approaches, that will make data from ground based studies more amenable to probabilistic risk analysis. While great progress has been made in determining the impact of space radiation on many advanced cognitive processes, challenges remain that need to be addressed prior to commencing deep space missions. A summary of on-going attempts to address existing knowledge gaps and the critical role that rodent studies will have in establishing the impact of space radiation on even more complex (human) cognitive tasks are presented and discussed.
Collapse
Affiliation(s)
- Richard A Britten
- Department of Radiation Oncology, Eastern Virginia Medical School, Norfolk, VA, 23507, USA; Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, 23507, USA; Leroy T Canoles Jr. Cancer Center, Eastern Virginia Medical School, Norfolk, VA, 23507, USA; Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA, 23507, USA.
| | - Laurie L Wellman
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA, 23507, USA; Department of Pathology & Anatomy, Eastern Virginia Medical School, Norfolk, VA, 23507, USA
| | - Larry D Sanford
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA, 23507, USA; Department of Pathology & Anatomy, Eastern Virginia Medical School, Norfolk, VA, 23507, USA
| |
Collapse
|
15
|
Sorokina SS, Malkov AE, Shubina LV, Zaichkina SI, Pikalov VA. Low dose of carbon ion irradiation induces early delayed cognitive impairments in mice. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2021; 60:61-71. [PMID: 33392787 DOI: 10.1007/s00411-020-00889-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 12/07/2020] [Indexed: 06/12/2023]
Abstract
People often encounter various sources of ionizing radiation, both in modern medicine and under various environmental conditions, such as space travel, nuclear power plants or in conditions of man-made disasters that may lead to long-term cognitive impairment. Whilst the effect of exposure to low and high doses of gamma and X-radiation on the central nervous system (CNS) has been well investigated, the consequences of protons and heavy ions irradiation are quite different and poorly understood. As for the assessment of long-term effects of carbon ions on cognitive abilities and neurodegeneration, very few data appeared in the literature. The main object of the research is to investigate the effects of accelerated carbon ions on the cognitive function. Experiments were performed on male SHK mice at an age of two months. Mice were irradiated with a dose of 0.7 Gy of accelerated carbon ions with an energy of 450 meV/n in spread-out Bragg peak (SOBP) on a U-70 particle accelerator (Protvino, Russia). Two months after the irradiation, mice were tested for total activity, spatial learning, as well as long- and short-term hippocampus-dependent memory. One month after the evaluation of cognitive activity, histological analysis of dorsal hippocampus was carried out to assess its morphological state and to reveal late neuronal degeneration. It was found that the mice irradiated with accelerated carbon ions develop an altered behavioral pattern characterized by anxiety and a shortage in hippocampal-dependent memory retention, but not in episodic memory. Nissl staining revealed a reduction in the number of cells in the dorsal hippocampus of irradiated mice, with the most pronounced reduction in cell density observed in the dentate gyrus (DG) hilus. Also, the length of the CA3 field of the dorsal hippocampus was significantly reduced, and the number of cells in it was moderately decreased. Experiments with the use of Fluoro-Jade B (FJB) staining revealed no FJB-positive regions in the dorsal hippocampus of irradiated and control animals 3 months after the irradiation. Thus, no morbid cells were detected in irradiated and control groups. The results obtained indicate that total irradiation with a low dose of carbon ions can produce a cognitive deficit in adult mice without evidence of neurodegenerative pathologic changes.
Collapse
Affiliation(s)
- S S Sorokina
- Institute of Theoretical and Experimental Biophysics Russian Academy of Sciences (ITEB RAS), Pushchino, Russia.
| | - A E Malkov
- Institute of Theoretical and Experimental Biophysics Russian Academy of Sciences (ITEB RAS), Pushchino, Russia
| | - L V Shubina
- Institute of Theoretical and Experimental Biophysics Russian Academy of Sciences (ITEB RAS), Pushchino, Russia
| | - S I Zaichkina
- Institute of Theoretical and Experimental Biophysics Russian Academy of Sciences (ITEB RAS), Pushchino, Russia
| | - V A Pikalov
- Institute of High Energy Physics Named by A.A. Logunov of National Research Centre "Kurchatov Institute", Protvino, Russia
| |
Collapse
|
16
|
Willey JS, Britten RA, Blaber E, Tahimic CG, Chancellor J, Mortreux M, Sanford LD, Kubik AJ, Delp MD, Mao XW. The individual and combined effects of spaceflight radiation and microgravity on biologic systems and functional outcomes. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART C, TOXICOLOGY AND CARCINOGENESIS 2021; 39:129-179. [PMID: 33902391 PMCID: PMC8274610 DOI: 10.1080/26896583.2021.1885283] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Both microgravity and radiation exposure in the spaceflight environment have been identified as hazards to astronaut health and performance. Substantial study has been focused on understanding the biology and risks associated with prolonged exposure to microgravity, and the hazards presented by radiation from galactic cosmic rays (GCR) and solar particle events (SPEs) outside of low earth orbit (LEO). To date, the majority of the ground-based analogues (e.g., rodent or cell culture studies) that investigate the biology of and risks associated with spaceflight hazards will focus on an individual hazard in isolation. However, astronauts will face these challenges simultaneously Combined hazard studies are necessary for understanding the risks astronauts face as they travel outside of LEO, and are also critical for countermeasure development. The focus of this review is to describe biologic and functional outcomes from ground-based analogue models for microgravity and radiation, specifically highlighting the combined effects of radiation and reduced weight-bearing from rodent ground-based tail suspension via hind limb unloading (HLU) and partial weight-bearing (PWB) models, although in vitro and spaceflight results are discussed as appropriate. The review focuses on the skeletal, ocular, central nervous system (CNS), cardiovascular, and stem cells responses.
Collapse
Affiliation(s)
| | | | - Elizabeth Blaber
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute
| | | | | | - Marie Mortreux
- Department of Neurology, Harvard Medical School, Beth Israel Deaconess Medical Center
| | - Larry D. Sanford
- Department of Radiation Oncology, Eastern Virginia Medical School
| | - Angela J. Kubik
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute
| | - Michael D. Delp
- Department of Nutrition, Food and Exercise Sciences, Florida State University
| | - Xiao Wen Mao
- Division of Biomedical Engineering Sciences (BMES), Department of Basic Sciences, Loma Linda University
| |
Collapse
|
17
|
Britten RA, Fesshaye AS, Duncan VD, Wellman LL, Sanford LD. Sleep Fragmentation Exacerbates Executive Function Impairments Induced by Low Doses of Si Ions. Radiat Res 2020; 194:116-123. [PMID: 32845991 DOI: 10.1667/rade-20-00080.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 05/22/2020] [Indexed: 11/03/2022]
Abstract
Astronauts on deep space missions will be required to work autonomously and thus their ability to perform executive functions could be critical to mission success. Ground-based rodent experiments have shown that low (<25 cGy) doses of several space radiation (SR) ions impair various aspects of executive function. Translating ground-based rodent studies into tangible risk estimates for astronauts remains an enormous challenge, but should similar neurocognitive impairments occur in astronauts exposed to low-SR doses, a Numbers-Needed-to-Harm analysis (of the rodent data) predicts that approximately 30% of the astronauts could develop severe cognitive flexibility decrements. In addition to the health risks associated with SR exposure, astronauts have to contend with other stressors, of which inadequate sleep quantity and quality are considered to be major concerns. We have shown that a single session of fragmented sleep uncovered latent attentional set-shifting (ATSET) performance deficits in rats exposed to protracted neutron radiation that had no obvious defects in performance under rested wakefulness conditions. It is unclear if the exacerbating effect of sleep fragmentation (SF) only occurs in rats receiving protracted low-dose-rate-neutron radiation. In this study, we assessed whether SF also unmasks latent ATSET deficits in rats exposed to 5 cGy 600 MeV/n 28Si ions. Only sham and Si-irradiated rats that had good ATSET performance (passing every stage of the test on their first attempt) were selected for study. Sleep fragmentation selectively impaired performance in the more complex IDR, EDS and EDR stages of the ATSET test in the Si-irradiated rats. Set-shifting performance has rarely been affected by SR exposure in our studies conducted with rats tested under rested wakefulness conditions. The consistent SF-related unmasking of latent set-shifting deficits in both Si- and neutron-irradiated rats suggests that there is a unique interaction between sleep fragmentation and space radiation on the functionality of the brain regions that regulate performance in the IDR, EDS and EDR stages of ATSET. The uncovering of these latent SR-induced ATSET performance deficits in both Si- and neutron-irradiated rats suggests that the true impact of SR-induced cognitive impairment may not be fully evident in normally rested rats, and thus cognitive testing needs to be conducted under both rested wakefulness and sleep fragmentation conditions.
Collapse
Affiliation(s)
- Richard A Britten
- Departments of a Radiation Oncology.,Departments of Microbiology and Molecular Cell Biology.,Center for Integrative Neuroscience and Inflammatory Diseases.,Leroy T. Canoles Jr. Cancer Center
| | | | | | - Laurie L Wellman
- Center for Integrative Neuroscience and Inflammatory Diseases.,Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Larry D Sanford
- Center for Integrative Neuroscience and Inflammatory Diseases.,Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, Virginia 23507
| |
Collapse
|
18
|
Franco-Pérez J, Montes S, Sánchez-Hernández J, Ballesteros-Zebadúa P. Whole-brain irradiation differentially modifies neurotransmitters levels and receptors in the hypothalamus and the prefrontal cortex. Radiat Oncol 2020; 15:269. [PMID: 33228731 PMCID: PMC7684903 DOI: 10.1186/s13014-020-01716-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 11/13/2020] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Whole-brain radiotherapy is a primary treatment for brain tumors and brain metastasis, but it also induces long-term undesired effects. Since cognitive impairment can occur, research on the etiology of secondary effects has focused on the hippocampus. Often overlooked, the hypothalamus controls critical homeostatic functions, some of which are also susceptible after whole-brain radiotherapy. Therefore, using whole-brain irradiation (WBI) in a rat model, we measured neurotransmitters and receptors in the hypothalamus. The prefrontal cortex and brainstem were also analyzed since they are highly connected to the hypothalamus and its regulatory processes. METHODS Male Wistar rats were exposed to WBI with 11 Gy (Biologically Effective Dose = 72 Gy). After 1 month, we evaluated changes in gamma-aminobutyric acid (GABA), glycine, taurine, aspartate, glutamate, and glutamine in the hypothalamus, prefrontal cortex, and brainstem according to an HPLC method. Ratios of Glutamate/GABA and Glutamine/Glutamate were calculated. Through Western Blott analysis, we measured the expression of GABAa and GABAb receptors, and NR1 and NR2A subunits of NMDA receptors. Changes were analyzed comparing results with sham controls using the non-parametric Mann-Whitney U test (p < 0.05). RESULTS WBI with 11 Gy induced significantly lower levels of GABA, glycine, taurine, aspartate, and GABAa receptor in the hypothalamus. Also, in the hypothalamus, a higher Glutamate/GABA ratio was found after irradiation. In the prefrontal cortex, WBI induced significant increases of glutamine and glutamate, Glutamine/Glutamate ratio, and increased expression of both GABAa receptor and NMDA receptor NR1 subunit. The brainstem showed no statistically significant changes after irradiation. CONCLUSION Our findings confirm that WBI can affect rat brain regions differently and opens new avenues for study. After 1 month, WBI decreases inhibitory neurotransmitters and receptors in the hypothalamus and, conversely, increases excitatory neurotransmitters and receptors in the prefrontal cortex. Increments in Glutamate/GABA in the hypothalamus and Glutamine/Glutamate in the frontal cortex indicate a neurochemical imbalance. Found changes could be related to several reported radiotherapy secondary effects, suggesting new prospects for therapeutic targets.
Collapse
Affiliation(s)
- Javier Franco-Pérez
- Laboratory of Physiology of Reticular Formation, National Institute of Neurology and Neurosurgery, INNN, Insurgentes Sur 3877, Col. La Fama, C.P. 14269, Mexico City, Mexico
| | - Sergio Montes
- Laboratory of Neurochemistry, National Institute of Neurology and Neurosurgery, INNN, Insurgentes Sur 3877, Col. La Fama, C.P. 14269, Mexico City, Mexico
| | - Josué Sánchez-Hernández
- Laboratory of Physiology of Reticular Formation, National Institute of Neurology and Neurosurgery, INNN, Insurgentes Sur 3877, Col. La Fama, C.P. 14269, Mexico City, Mexico
| | - Paola Ballesteros-Zebadúa
- Laboratory of Medical Physics, National Institute of Neurology and Neurosurgery, INNN, Insurgentes Sur 3877, Col. La Fama, C.P. 14269, Mexico City, Mexico.
| |
Collapse
|
19
|
Blackwell AA, Schell BD, Osterlund Oltmanns JR, Whishaw IQ, Ton ST, Adamczyk NS, Kartje GL, Britten RA, Wallace DG. Skilled movement and posture deficits in rat string-pulling behavior following low dose space radiation ( 28Si) exposure. Behav Brain Res 2020; 400:113010. [PMID: 33181183 DOI: 10.1016/j.bbr.2020.113010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 10/21/2020] [Accepted: 11/05/2020] [Indexed: 10/23/2022]
Abstract
Deep space flight missions beyond the Van Allen belt have the potential to expose astronauts to space radiation which may damage the central nervous system and impair function. The proposed mission to Mars will be the longest mission-to-date and identifying mission critical tasks that are sensitive to space radiation is important for developing and evaluating the efficacy of counter measures. Fine motor control has been assessed in humans, rats, and many other species using string-pulling behavior. For example, focal cortical damage has been previously shown to disrupt the topographic (i.e., path circuity) and kinematic (i.e., moment-to-moment speed) organization of rat string-pulling behavior count to compromise task accuracy. In the current study, rats were exposed to a ground-based model of simulated space radiation (5 cGy 28Silicon), and string-pulling behavior was used to assess fine motor control. Irradiated rats initially took longer to pull an unweighted string into a cage, exhibited impaired accuracy in grasping the string, and displayed postural deficits. Once rats were switched to a weighted string, some deficits lessened but postural instability remained. These results demonstrate that a single exposure to a low dose of space radiation disrupts skilled hand movements and posture, suggestive of neural impairment. This work establishes a foundation for future studies to investigate the neural structures and circuits involved in fine motor control and to examine the effectiveness of counter measures to attenuate the effects of space radiation on fine motor control.
Collapse
Affiliation(s)
- Ashley A Blackwell
- Department of Psychology, Northern Illinois University, DeKalb, IL, 60115, United States.
| | - Brandi D Schell
- Department of Psychology, Northern Illinois University, DeKalb, IL, 60115, United States
| | | | - Ian Q Whishaw
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, Alberta, Canada
| | - Son T Ton
- Research Service, Edward Hines Jr. VA Hospital, Hines, IL, 60141, United States; Department of Molecular Pharmacology and Neuroscience, Loyola University Chicago Health Sciences Division, Maywood, IL, 60153, United States
| | - Natalie S Adamczyk
- Research Service, Edward Hines Jr. VA Hospital, Hines, IL, 60141, United States
| | - Gwendolyn L Kartje
- Research Service, Edward Hines Jr. VA Hospital, Hines, IL, 60141, United States; Department of Molecular Pharmacology and Neuroscience, Loyola University Chicago Health Sciences Division, Maywood, IL, 60153, United States
| | - Richard A Britten
- Department of Radiation Oncology, Eastern Virginia Medical School, Norfolk, VA, 23507, United States
| | - Douglas G Wallace
- Department of Psychology, Northern Illinois University, DeKalb, IL, 60115, United States
| |
Collapse
|
20
|
Pariset E, Malkani S, Cekanaviciute E, Costes SV. Ionizing radiation-induced risks to the central nervous system and countermeasures in cellular and rodent models. Int J Radiat Biol 2020; 97:S132-S150. [PMID: 32946305 DOI: 10.1080/09553002.2020.1820598] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE Harmful effects of ionizing radiation on the Central Nervous System (CNS) are a concerning outcome in the field of cancer radiotherapy and form a major risk for deep space exploration. Both acute and chronic CNS irradiation induce a complex network of molecular and cellular alterations including DNA damage, oxidative stress, cell death and systemic inflammation, leading to changes in neuronal structure and synaptic plasticity with behavioral and cognitive consequences in animal models. Due to this complexity, countermeasure or therapeutic approaches to reduce the harmful effects of ionizing radiation include a wide range of protective and mitigative strategies, which merit a thorough comparative analysis. MATERIALS AND METHODS We reviewed current approaches for developing countermeasures to both targeted and non-targeted effects of ionizing radiation on the CNS from the molecular and cellular to the behavioral level. RESULTS We focus on countermeasures that aim to mitigate the four main detrimental actions of radiation on CNS: DNA damage, free radical formation and oxidative stress, cell death, and harmful systemic responses including tissue death and neuroinflammation. We propose a comprehensive review of CNS radiation countermeasures reported for the full range of irradiation types (photons and particles, low and high linear energy transfer) and doses (from a fraction of gray to several tens of gray, fractionated and unfractionated), with a particular interest for exposure conditions relevant to deep-space environment and radiotherapy. Our review reveals the importance of combined strategies that increase DNA protection and repair, reduce free radical formation and increase their elimination, limit inflammation and improve cell viability, limit tissue damage and increase repair and plasticity. CONCLUSIONS The majority of therapeutic approaches to protect the CNS from ionizing radiation have been limited to acute high dose and high dose rate gamma irradiation, and few are translatable from animal models to potential human application due to harmful side effects and lack of blood-brain barrier permeability that precludes peripheral administration. Therefore, a promising research direction would be to focus on practical applicability and effectiveness in a wider range of irradiation paradigms, from fractionated therapeutic to deep space radiation. In addition to discovering novel therapeutics, it would be worth maximizing the benefits and reducing side effects of those that already exist. Finally, we suggest that novel cellular and tissue models for developing and testing countermeasures in the context of other impairments might also be applied to the field of CNS responses to ionizing radiation.
Collapse
Affiliation(s)
- Eloise Pariset
- Universities Space Research Association, Columbia, MD, USA.,Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
| | - Sherina Malkani
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA.,Young Scientist Program, Blue Marble Space Institute of Science, Moffett Field, CA, USA
| | - Egle Cekanaviciute
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
| | - Sylvain V Costes
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
| |
Collapse
|
21
|
Clément GR, Boyle RD, George KA, Nelson GA, Reschke MF, Williams TJ, Paloski WH. Challenges to the central nervous system during human spaceflight missions to Mars. J Neurophysiol 2020; 123:2037-2063. [DOI: 10.1152/jn.00476.2019] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Space travel presents a number of environmental challenges to the central nervous system, including changes in gravitational acceleration that alter the terrestrial synergies between perception and action, galactic cosmic radiation that can damage sensitive neurons and structures, and multiple factors (isolation, confinement, altered atmosphere, and mission parameters, including distance from Earth) that can affect cognition and behavior. Travelers to Mars will be exposed to these environmental challenges for up to 3 years, and space-faring nations continue to direct vigorous research investments to help elucidate and mitigate the consequences of these long-duration exposures. This article reviews the findings of more than 50 years of space-related neuroscience research on humans and animals exposed to spaceflight or analogs of spaceflight environments, and projects the implications and the forward work necessary to ensure successful Mars missions. It also reviews fundamental neurophysiology responses that will help us understand and maintain human health and performance on Earth.
Collapse
Affiliation(s)
| | - Richard D. Boyle
- National Aeronautics and Space Administration, Ames Research Center, Moffett Field, California
| | | | - Gregory A. Nelson
- Division of Biomedical Engineering Sciences, School of Medicine Loma Linda University, Loma Linda, California
| | - Millard F. Reschke
- National Aeronautics and Space Administration, Johnson Space Center, Houston, Texas
| | - Thomas J. Williams
- National Aeronautics and Space Administration, Johnson Space Center, Houston, Texas
| | - William H. Paloski
- National Aeronautics and Space Administration, Johnson Space Center, Houston, Texas
| |
Collapse
|
22
|
Mayer M, Arrizabalaga O, Ciba M, Schroeder IS, Ritter S, Thielemann C. Novel in vitro assay to investigate radiation induced changes in the functionality of human embryonic stem cell-derived neurospheres. Neurotoxicology 2020; 79:40-47. [PMID: 32320710 DOI: 10.1016/j.neuro.2020.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 03/31/2020] [Accepted: 04/15/2020] [Indexed: 10/24/2022]
Abstract
Ionizing radiation (IR) is increasingly used for diagnostics and therapy of severe brain diseases. However, IR also has adverse effects on the healthy brain tissue, particularly on the neuronal network. This is true for adults but even more pronounced in the developing brain of unborn and pediatric patients. Epidemiological studies on children receiving radiotherapy showed an increased risk for cognitive decline ranging from mild deficits in academic functioning to severe late effects in intellectual ability and language as a consequence of altered neuronal development and connectivity. To provide a comprehensive approach for the analysis of radiation-induced alterations in human neuronal functionality, we developed an in vitro assay by combining microelectrode array (MEA) analyses and human embryonic stem cell (hESC) derived three-dimensional neurospheres (NS). In our proof of principle study, we irradiated hESC with 1 Gy X-rays and let them spontaneously differentiate into neurons within NS. After the onset of neuronal activity, we recorded and analyzed the activity pattern of the developing neuronal networks. The network activity in NS derived from irradiated hESC was significantly reduced, whereas no differences in molecular endpoints such as cell proliferation and transcript or protein expression analyses were found. Thus, the combination of MEA analysis with a 3D model for neuronal functionality revealed radiation sequela that otherwise would not have been detected. We therefore strongly suggest combining traditional biomolecular methods with the new functional assay presented in this work to improve the risk assessment for IR-induced effects on the developing brain.
Collapse
Affiliation(s)
- Margot Mayer
- TH Aschaffenburg University of Applied Sciences, BioMEMS Lab, Aschaffenburg, Germany.
| | - Onetsine Arrizabalaga
- GSI Helmholtzzentrum für Schwerionenforschung, Biophysics Division, Darmstadt, Germany.
| | - Manuel Ciba
- TH Aschaffenburg University of Applied Sciences, BioMEMS Lab, Aschaffenburg, Germany.
| | - Insa S Schroeder
- GSI Helmholtzzentrum für Schwerionenforschung, Biophysics Division, Darmstadt, Germany.
| | - Sylvia Ritter
- GSI Helmholtzzentrum für Schwerionenforschung, Biophysics Division, Darmstadt, Germany.
| | - Christiane Thielemann
- TH Aschaffenburg University of Applied Sciences, BioMEMS Lab, Aschaffenburg, Germany.
| |
Collapse
|
23
|
Whoolery CW, Yun S, Reynolds RP, Lucero MJ, Soler I, Tran FH, Ito N, Redfield RL, Richardson DR, Shih HY, Rivera PD, Chen BPC, Birnbaum SG, Stowe AM, Eisch AJ. Multi-domain cognitive assessment of male mice shows space radiation is not harmful to high-level cognition and actually improves pattern separation. Sci Rep 2020; 10:2737. [PMID: 32066765 PMCID: PMC7026431 DOI: 10.1038/s41598-020-59419-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 01/23/2020] [Indexed: 12/20/2022] Open
Abstract
Astronauts on interplanetary missions - such as to Mars - will be exposed to space radiation, a spectrum of highly-charged, fast-moving particles that includes 56Fe and 28Si. Earth-based preclinical studies show space radiation decreases rodent performance in low- and some high-level cognitive tasks. Given astronaut use of touchscreen platforms during training and space flight and given the ability of rodent touchscreen tasks to assess functional integrity of brain circuits and multiple cognitive domains in a non-aversive way, here we exposed 6-month-old C57BL/6J male mice to whole-body space radiation and subsequently assessed them on a touchscreen battery. Relative to Sham treatment, 56Fe irradiation did not overtly change performance on tasks of visual discrimination, reversal learning, rule-based, or object-spatial paired associates learning, suggesting preserved functional integrity of supporting brain circuits. Surprisingly, 56Fe irradiation improved performance on a dentate gyrus-reliant pattern separation task; irradiated mice learned faster and were more accurate than controls. Improved pattern separation performance did not appear to be touchscreen-, radiation particle-, or neurogenesis-dependent, as 56Fe and 28Si irradiation led to faster context discrimination in a non-touchscreen task and 56Fe decreased new dentate gyrus neurons relative to Sham. These data urge revisitation of the broadly-held view that space radiation is detrimental to cognition.
Collapse
Affiliation(s)
- Cody W Whoolery
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Sanghee Yun
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Anesthesiology and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ryan P Reynolds
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Melanie J Lucero
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ivan Soler
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Fionya H Tran
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Naoki Ito
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Oriental Medicine Research Center, Kitasato University, Tokyo, Japan
| | - Rachel L Redfield
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Devon R Richardson
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Hung-Ying Shih
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Phillip D Rivera
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Biology, Hope College, Holland, MI, USA
| | - Benjamin P C Chen
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Shari G Birnbaum
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ann M Stowe
- Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, Dallas, TX, USA
- Department of Neurology, University of Kentucky, Lexington, KY, USA
| | - Amelia J Eisch
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Anesthesiology and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
24
|
Britten RA, Duncan VD, Fesshaye A, Rudobeck E, Nelson GA, Vlkolinsky R. Altered Cognitive Flexibility and Synaptic Plasticity in the Rat Prefrontal Cortex after Exposure to Low (≤15 cGy) Doses of 28Si Radiation. Radiat Res 2020; 193:223-235. [DOI: 10.1667/rr15458.1] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
| | | | | | - Emil Rudobeck
- Department of Basic Sciences, Loma Linda University, Loma Linda, California, 92354
| | - Gregory A. Nelson
- Department of Basic Sciences, Loma Linda University, Loma Linda, California, 92354
| | - Roman Vlkolinsky
- Department of Basic Sciences, Loma Linda University, Loma Linda, California, 92354
| |
Collapse
|
25
|
Kokhan VS, Anokhin PK, Belov OV, Gulyaev MV. Cortical Glutamate/GABA Imbalance after Combined Radiation Exposure: Relevance to Human Deep-Space Missions. Neuroscience 2019; 416:295-308. [DOI: 10.1016/j.neuroscience.2019.08.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 07/01/2019] [Accepted: 08/03/2019] [Indexed: 12/22/2022]
|
26
|
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: 34] [Impact Index Per Article: 6.8] [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
|
27
|
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: 10.8] [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
|
28
|
Whole-Body 12C Irradiation Transiently Decreases Mouse Hippocampal Dentate Gyrus Proliferation and Immature Neuron Number, but Does Not Change New Neuron Survival Rate. Int J Mol Sci 2018; 19:ijms19103078. [PMID: 30304778 PMCID: PMC6213859 DOI: 10.3390/ijms19103078] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 09/17/2018] [Accepted: 09/21/2018] [Indexed: 02/08/2023] Open
Abstract
High-charge and -energy (HZE) particles comprise space radiation and they pose a challenge to astronauts on deep space missions. While exposure to most HZE particles decreases neurogenesis in the hippocampus—a brain structure important in memory—prior work suggests that 12C does not. However, much about 12C’s influence on neurogenesis remains unknown, including the time course of its impact on neurogenesis. To address this knowledge gap, male mice (9–11 weeks of age) were exposed to whole-body 12C irradiation 100 cGy (IRR; 1000 MeV/n; 8 kEV/µm) or Sham treatment. To birthdate dividing cells, mice received BrdU i.p. 22 h post-irradiation and brains were harvested 2 h (Short-Term) or three months (Long-Term) later for stereological analysis indices of dentate gyrus neurogenesis. For the Short-Term time point, IRR mice had fewer Ki67, BrdU, and doublecortin (DCX) immunoreactive (+) cells versus Sham mice, indicating decreased proliferation (Ki67, BrdU) and immature neurons (DCX). For the Long-Term time point, IRR and Sham mice had similar Ki67+ and DCX+ cell numbers, suggesting restoration of proliferation and immature neurons 3 months post-12C irradiation. IRR mice had fewer surviving BrdU+ cells versus Sham mice, suggesting decreased cell survival, but there was no difference in BrdU+ cell survival rate when compared within treatment and across time point. These data underscore the ability of neurogenesis in the mouse brain to recover from the detrimental effect of 12C exposure.
Collapse
|
29
|
Hellweg CE, Chishti AA, Diegeler S, Spitta LF, Henschenmacher B, Baumstark-Khan C. Molecular Signaling in Response to Charged Particle Exposures and its Importance in Particle Therapy. Int J Part Ther 2018; 5:60-73. [PMID: 31773020 PMCID: PMC6871585 DOI: 10.14338/ijpt-18-00016.1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 06/13/2018] [Indexed: 12/17/2022] Open
Abstract
Energetic, charged particles elicit an orchestrated DNA damage response (DDR) during their traversal through healthy tissues and tumors. Complex DNA damage formation, after exposure to high linear energy transfer (LET) charged particles, results in DNA repair foci formation, which begins within seconds. More protein modifications occur after high-LET, compared with low-LET, irradiation. Charged-particle exposure activates several transcription factors that are cytoprotective or cytodestructive, or that upregulate cytokine and chemokine expression, and are involved in bystander signaling. Molecular signaling for a survival or death decision in different tumor types and healthy tissues should be studied as prerequisite for shaping sensitizing and protective strategies. Long-term signaling and gene expression changes were found in various tissues of animals exposed to charged particles, and elucidation of their role in chronic and late effects of charged-particle therapy will help to develop effective preventive measures.
Collapse
Affiliation(s)
- Christine E. Hellweg
- German Aerospace Center (DLR), Institute of Aerospace Medicine, Radiation Biology Department, Köln, Germany
| | - Arif Ali Chishti
- German Aerospace Center (DLR), Institute of Aerospace Medicine, Radiation Biology Department, Köln, Germany
- The Karachi Institute of Biotechnology and Genetic Engineering, University of Karachi, Karachi, Pakistan
| | - Sebastian Diegeler
- German Aerospace Center (DLR), Institute of Aerospace Medicine, Radiation Biology Department, Köln, Germany
| | - Luis F. Spitta
- German Aerospace Center (DLR), Institute of Aerospace Medicine, Radiation Biology Department, Köln, Germany
| | - Bernd Henschenmacher
- German Aerospace Center (DLR), Institute of Aerospace Medicine, Radiation Biology Department, Köln, Germany
| | - Christa Baumstark-Khan
- German Aerospace Center (DLR), Institute of Aerospace Medicine, Radiation Biology Department, Köln, Germany
| |
Collapse
|
30
|
Britten RA, Jewell JS, Duncan VD, Hadley MM, Macadat E, Musto AE, Tessa CL. Impaired Attentional Set-Shifting Performance after Exposure to 5 cGy of 600 MeV/n28Si Particles. Radiat Res 2018; 189:273-282. [DOI: 10.1667/rr14627.1] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Richard A. Britten
- Department of a 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
| | - Jessica S. Jewell
- Department of a Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Vania D. Duncan
- Department of a Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Melissa M. Hadley
- Department of a Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Evangeline Macadat
- Department of a Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Alberto E. Musto
- Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Chiara La Tessa
- NSRL Brookhaven National Laboratories, Upton, New York 11973
- University of Trento, Povo Trento 38122, Italy
| |
Collapse
|
31
|
Ji S, Ding X, Ji J, Wu H, Sun R, Li X, Zhang L, Tian Y. Cranial irradiation inhibits hippocampal neurogenesis via DNMT1 and DNMT3A. Oncol Lett 2018; 15:2899-2904. [PMID: 29435016 PMCID: PMC5778827 DOI: 10.3892/ol.2017.7643] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 11/16/2017] [Indexed: 02/07/2023] Open
Abstract
Impairment of neurogenesis in the hippocampus following whole-brain irradiation is the most important mechanism of radiation-induced cognitive dysfunction. However, the underlying mechanism remains obscure, meaning an ideal therapeutic target has not been identified. Evidence indicates that DNA methylation in neurons regulates synaptic plasticity and neuronal network activity. In the present study, the expression of DNA methyltransferases (DNMTs) in the hippocampus was analyzed to investigate their potential function in radiation-induced neurogenesis impairment. Sprague-Dawley rats were used throughout the present study, apportioned to the following groups: Control, radiation only, zebularine (a DNMT inhibitor) only, and radiation and zebularine together. Immunofluorescence staining revealed that radiation inhibited cellular proliferation and dendritic growth within new neurons of the hippocampus. In addition, western blot analysis demonstrated lower expression levels of DNMT1 and DNMT3A protein following radiation treatment compared with that in the non-irradiated control. Furthermore, compared with the radiation-only group, the radiation and zebularine group had significantly lower cell proliferative abilities, dendritic growth, and DNMT1 and DNMT3A protein levels. The results of the present study indicated that DNMT1 and DNMT3A may be involved in the pathogenesis of whole-brain radiation-induced neurogenesis impairment.
Collapse
Affiliation(s)
- Shengjun Ji
- Cancer Center, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, Jiangsu 215001, P.R. China
| | - Xin Ding
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Jiang Ji
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Haohao Wu
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Rui Sun
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Xiaoyang Li
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Liyuan Zhang
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Ye Tian
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| |
Collapse
|
32
|
Rudobeck E, Bellone JA, Szücs A, Bonnick K, Mehrotra-Carter S, Badaut J, Nelson GA, Hartman RE, Vlkolinský R. Low-dose proton radiation effects in a transgenic mouse model of Alzheimer's disease - Implications for space travel. PLoS One 2017; 12:e0186168. [PMID: 29186131 PMCID: PMC5706673 DOI: 10.1371/journal.pone.0186168] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 09/26/2017] [Indexed: 12/13/2022] Open
Abstract
Space radiation represents a significant health risk for astronauts. Ground-based animal studies indicate that space radiation affects neuronal functions such as excitability, synaptic transmission, and plasticity, and it may accelerate the onset of Alzheimer's disease (AD). Although protons represent the main constituent in the space radiation spectrum, their effects on AD-related pathology have not been tested. We irradiated 3 month-old APP/PSEN1 transgenic (TG) and wild type (WT) mice with protons (150 MeV; 0.1-1.0 Gy; whole body) and evaluated functional and biochemical hallmarks of AD. We performed behavioral tests in the water maze (WM) before irradiation and in the WM and Barnes maze at 3 and 6 months post-irradiation to evaluate spatial learning and memory. We also performed electrophysiological recordings in vitro in hippocampal slices prepared 6 and 9 months post-irradiation to evaluate excitatory synaptic transmission and plasticity. Next, we evaluated amyloid β (Aβ) deposition in the contralateral hippocampus and adjacent cortex using immunohistochemistry. In cortical homogenates, we analyzed the levels of the presynaptic marker synaptophysin by Western blotting and measured pro-inflammatory cytokine levels (TNFα, IL-1β, IL-6, CXCL10 and CCL2) by bead-based multiplex assay. TG mice performed significantly worse than WT mice in the WM. Irradiation of TG mice did not affect their behavioral performance, but reduced the amplitudes of population spikes and inhibited paired-pulse facilitation in CA1 neurons. These electrophysiological alterations in the TG mice were qualitatively different from those observed in WT mice, in which irradiation increased excitability and synaptic efficacy. Irradiation increased Aβ deposition in the cortex of TG mice without affecting cytokine levels and increased synaptophysin expression in WT mice (but not in the TG mice). Although irradiation with protons increased Aβ deposition, the complex functional and biochemical results indicate that irradiation effects are not synergistic to AD pathology.
Collapse
Affiliation(s)
- Emil Rudobeck
- Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, United States of America
| | - John A. Bellone
- Department of Psychology, School of Behavioral Health, Loma Linda University, Loma Linda, CA, United States of America
| | - Attila Szücs
- BioCircuits Institute, University of California San Diego, La Jolla, CA, United States of America
| | - Kristine Bonnick
- Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, United States of America
| | - Shalini Mehrotra-Carter
- Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, United States of America
| | - Jerome Badaut
- Department of Physiology, School of Medicine, Loma Linda University, Loma Linda, CA, United States of America
| | - Gregory A. Nelson
- Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, United States of America
| | - Richard E. Hartman
- Department of Psychology, School of Behavioral Health, Loma Linda University, Loma Linda, CA, United States of America
| | - Roman Vlkolinský
- Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, United States of America
| |
Collapse
|
33
|
Whoolery CW, Walker AK, Richardson DR, Lucero MJ, Reynolds RP, Beddow DH, Clark KL, Shih HY, LeBlanc JA, Cole MG, Amaral WZ, Mukherjee S, Zhang S, Ahn F, Bulin SE, DeCarolis NA, Rivera PD, Chen BPC, Yun S, Eisch AJ. Whole-Body Exposure to 28Si-Radiation Dose-Dependently Disrupts Dentate Gyrus Neurogenesis and Proliferation in the Short Term and New Neuron Survival and Contextual Fear Conditioning in the Long Term. Radiat Res 2017; 188:532-551. [PMID: 28945526 PMCID: PMC5901735 DOI: 10.1667/rr14797.1] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Astronauts traveling to Mars will be exposed to chronic low doses of galactic cosmic space radiation, which contains highly charged, high-energy (HZE) particles. 56Fe-HZE-particle exposure decreases hippocampal dentate gyrus (DG) neurogenesis and disrupts hippocampal function in young adult rodents, raising the possibility of impaired astronaut cognition and risk of mission failure. However, far less is known about how exposure to other HZE particles, such as 28Si, influences hippocampal neurogenesis and function. To compare the influence of 28Si exposure on indices of neurogenesis and hippocampal function with previous studies on 56Fe exposure, 9-week-old C57BL/6J and Nestin-GFP mice (NGFP; made and maintained for 10 or more generations on a C57BL/6J background) received whole-body 28Si-particle-radiation exposure (0, 0.2 and 1 Gy, 300 MeV/n, LET 67 KeV/μ, dose rate 1 Gy/min). For neurogenesis assessment, the NGFP mice were injected with the mitotic marker BrdU at 22 h postirradiation and brains were examined for indices of hippocampal proliferation and neurogenesis, including Ki67+, BrdU+, BrdU+NeuN+ and DCX+ cell numbers at short- and long-term time points (24 h and 3 months postirradiation, respectively). In the short-term group, stereology revealed fewer Ki67+, BrdU+ and DCX+ cells in 1-Gy-irradiated group relative to nonirradiated control mice, fewer Ki67+ and DCX+ cells in 0.2 Gy group relative to control group and fewer BrdU+ and DCX+ cells in 1 Gy group relative to 0.2 Gy group. In contrast to the clearly observed radiation-induced, dose-dependent reductions in the short-term group across all markers, only a few neurogenesis indices were changed in the long-term irradiated groups. Notably, there were fewer surviving BrdU+ cells in the 1 Gy group relative to 0- and 0.2-Gy-irradiated mice in the long-term group. When the short- and long-term groups were analyzed by sex, exposure to radiation had a similar effect on neurogenesis indices in male and female mice, although only male mice showed fewer surviving BrdU+ cells in the long-term group. Fluorescent immunolabeling and confocal phenotypic analysis revealed that most surviving BrdU+ cells in the long-term group expressed the neuronal marker NeuN, definitively confirming that exposure to 1 Gy 28Si radiation decreased the number of surviving adult-generated neurons in male mice relative to both 0- and 0.2-Gy-irradiated mice. For hippocampal function assessment, 9-week-old male C57BL/6J mice received whole-body 28Si-particle exposure and were then assessed long-term for performance on contextual and cued fear conditioning. In the context test the animals that received 0.2 Gy froze less relative to control animals, suggesting decreased hippocampal-dependent function. However, in the cued fear conditioning test, animals that received 1 Gy froze more during the pretone portion of the test, relative to controls and 0.2-Gy-irradiated mice, suggesting enhanced anxiety. Compared to previously reported studies, these data suggest that 28Si-radiation exposure damages neurogenesis, but to a lesser extent than 56Fe radiation and that low-dose 28Si exposure induces abnormalities in hippocampal function, disrupting fear memory but also inducing anxiety-like behavior. Furthermore, exposure to 28Si radiation decreased new neuron survival in long-term male groups but not females suggests that sex may be an important factor when performing brain health risk assessment for astronauts traveling in space.
Collapse
Affiliation(s)
- Cody W. Whoolery
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, Texas
| | - Angela K. Walker
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, Texas
| | | | - Melanie J. Lucero
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, Texas
| | - Ryan P. Reynolds
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, Texas
- Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - David H. Beddow
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, Texas
| | - K. Lyles Clark
- Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Hung-Ying Shih
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, Texas
| | - Junie A. LeBlanc
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, Texas
| | - Mara G. Cole
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, Texas
| | | | - Shibani Mukherjee
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, Texas
| | - Shichuan Zhang
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, Texas
| | - Francisca Ahn
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, Texas
| | - Sarah E. Bulin
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, Texas
| | | | - Phillip D. Rivera
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, Texas
| | - Benjamin P. C. Chen
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, Texas
| | - Sanghee Yun
- Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Amelia J. Eisch
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, Texas
- Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| |
Collapse
|
34
|
Lee SH, Dudok B, Parihar VK, Jung KM, Zöldi M, Kang YJ, Maroso M, Alexander AL, Nelson GA, Piomelli D, Katona I, Limoli CL, Soltesz I. Neurophysiology of space travel: energetic solar particles cause cell type-specific plasticity of neurotransmission. Brain Struct Funct 2016; 222:2345-2357. [PMID: 27905022 PMCID: PMC5504243 DOI: 10.1007/s00429-016-1345-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 11/25/2016] [Indexed: 12/03/2022]
Abstract
In the not too distant future, humankind will embark on one of its greatest adventures, the travel to distant planets. However, deep space travel is associated with an inevitable exposure to radiation fields. Space-relevant doses of protons elicit persistent disruptions in cognition and neuronal structure. However, whether space-relevant irradiation alters neurotransmission is unknown. Within the hippocampus, a brain region crucial for cognition, perisomatic inhibitory control of pyramidal cells (PCs) is supplied by two distinct cell types, the cannabinoid type 1 receptor (CB1)-expressing basket cells (CB1BCs) and parvalbumin (PV)-expressing interneurons (PVINs). Mice subjected to low-dose proton irradiation were analyzed using electrophysiological, biochemical and imaging techniques months after exposure. In irradiated mice, GABA release from CB1BCs onto PCs was dramatically increased. This effect was abolished by CB1 blockade, indicating that irradiation decreased CB1-dependent tonic inhibition of GABA release. These alterations in GABA release were accompanied by decreased levels of the major CB1 ligand 2-arachidonoylglycerol. In contrast, GABA release from PVINs was unchanged, and the excitatory connectivity from PCs to the interneurons also underwent cell type-specific alterations. These results demonstrate that energetic charged particles at space-relevant low doses elicit surprisingly selective long-term plasticity of synaptic microcircuits in the hippocampus. The magnitude and persistent nature of these alterations in synaptic function are consistent with the observed perturbations in cognitive performance after irradiation, while the high specificity of these changes indicates that it may be possible to develop targeted therapeutic interventions to decrease the risk of adverse events during interplanetary travel.
Collapse
Affiliation(s)
- Sang-Hun Lee
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, 92697, USA. .,Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA.
| | - Barna Dudok
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, 1083, Budapest, Hungary.,School of Ph.D. Studies, Semmelweis University, Budapest, Hungary
| | - Vipan K Parihar
- Department of Radiation Oncology, University of California, Irvine, CA, 92697, USA
| | - Kwang-Mook Jung
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, 92697, USA
| | - Miklós Zöldi
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, 1083, Budapest, Hungary.,School of Ph.D. Studies, Semmelweis University, Budapest, Hungary
| | - Young-Jin Kang
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Mattia Maroso
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, 92697, USA.,Department of Neurosurgery, and Neurology and Neurological Sciences, Stanford University, Palo Alto, CA, 94305, USA
| | - Allyson L Alexander
- Department of Neurosurgery, and Neurology and Neurological Sciences, Stanford University, Palo Alto, CA, 94305, USA
| | - Gregory A Nelson
- Division of Radiation Research, Department of Basic Sciences, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Daniele Piomelli
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, 92697, USA
| | - István Katona
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, 1083, Budapest, Hungary
| | - Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine, CA, 92697, USA
| | - Ivan Soltesz
- Department of Neurosurgery, and Neurology and Neurological Sciences, Stanford University, Palo Alto, CA, 94305, USA
| |
Collapse
|
35
|
Hadley MM, Davis LK, Jewell JS, Miller VD, Britten RA. Exposure to Mission-Relevant Doses of 1 GeV/n48Ti Particles Impairs Attentional Set-Shifting Performance in Retired Breeder Rats. Radiat Res 2016; 185:13-9. [DOI: 10.1667/rr14086.1] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
36
|
Raber J, Allen AR, Sharma S, Allen B, Rosi S, Olsen RHJ, Davis MJ, Eiwaz M, Fike JR, Nelson GA. Effects of Proton and Combined Proton and 56Fe Radiation on the Hippocampus. Radiat Res 2015; 185:20-30. [DOI: 10.1667/rr14222.1] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
| | - Antiño R. Allen
- Brain and Spinal Injury Center, Department of Neurological Surgery,
| | - Sourabh Sharma
- Brain and Spinal Injury Center, Department of Neurological Surgery,
| | - Barrett Allen
- Brain and Spinal Injury Center, Department of Neurological Surgery,
| | - Susanna Rosi
- Brain and Spinal Injury Center, Department of Neurological Surgery,
| | | | | | | | - John R. Fike
- Brain and Spinal Injury Center, Department of Neurological Surgery,
| | - Gregory A. Nelson
- Department of Basic Sciences, Division of Radiation Research, Loma Linda University, Loma Linda, California, 92350
| |
Collapse
|
37
|
Alp M, Parihar VK, Limoli CL, Cucinotta FA. Irradiation of Neurons with High-Energy Charged Particles: An In Silico Modeling Approach. PLoS Comput Biol 2015; 11:e1004428. [PMID: 26252394 PMCID: PMC4529238 DOI: 10.1371/journal.pcbi.1004428] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Accepted: 07/03/2015] [Indexed: 11/19/2022] Open
Abstract
In this work, a stochastic computational model of microscopic energy deposition events is used to study for the first time damage to irradiated neuronal cells of the mouse hippocampus. An extensive library of radiation tracks for different particle types is created to score energy deposition in small voxels and volume segments describing a neuron's morphology that later are sampled for given particle fluence or dose. Methods included the construction of in silico mouse hippocampal granule cells from neuromorpho.org with spine and filopodia segments stochastically distributed along the dendritic branches. The model is tested with high-energy (56)Fe, (12)C, and (1)H particles and electrons. Results indicate that the tree-like structure of the neuronal morphology and the microscopic dose deposition of distinct particles may lead to different outcomes when cellular injury is assessed, leading to differences in structural damage for the same absorbed dose. The significance of the microscopic dose in neuron components is to introduce specific local and global modes of cellular injury that likely contribute to spine, filopodia, and dendrite pruning, impacting cognition and possibly the collapse of the neuron. Results show that the heterogeneity of heavy particle tracks at low doses, compared to the more uniform dose distribution of electrons, juxtaposed with neuron morphology make it necessary to model the spatial dose painting for specific neuronal components. Going forward, this work can directly support the development of biophysical models of the modifications of spine and dendritic morphology observed after low dose charged particle irradiation by providing accurate descriptions of the underlying physical insults to complex neuron structures at the nano-meter scale.
Collapse
Affiliation(s)
- Murat Alp
- Department of Health Physics and Diagnostic Sciences, University of Nevada, Las Vegas, Las Vegas, Nevada, United States of America
| | - Vipan K. Parihar
- Department of Radiation Oncology, University of California, Irvine, Irvine, California, United States of America
| | - Charles L. Limoli
- Department of Radiation Oncology, University of California, Irvine, Irvine, California, United States of America
| | - Francis A. Cucinotta
- Department of Health Physics and Diagnostic Sciences, University of Nevada, Las Vegas, Las Vegas, Nevada, United States of America
| |
Collapse
|
38
|
Bellone JA, Rudobeck E, Hartman RE, Szücs A, Vlkolinský R. A Single Low Dose of Proton Radiation Induces Long-Term Behavioral and Electrophysiological Changes in Mice. Radiat Res 2015. [DOI: 10.1667/rr13903.1] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|