1
|
Kumar R, Kumari P, Kumar R. Central Nervous System Response Against Ionizing Radiation Exposure: Cellular, Biochemical, and Molecular Perspectives. Mol Neurobiol 2025; 62:7268-7295. [PMID: 39875779 DOI: 10.1007/s12035-025-04712-z] [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: 07/11/2024] [Accepted: 01/15/2025] [Indexed: 01/30/2025]
Abstract
Gamma radiation is known to induce several detrimental effects on the nervous system. The hippocampus region, specifically the dentate gyrus (DG) and subventricular zone (SVZ), have been identified as a radiation-sensitive neurogenic niche. Radiation alters the endogenous redox status of neural stem cells (NSCs) and other proliferative cells, especially in the hippocampus region, leading to oxidative stress, neuroinflammation, and cell death. Planned (i.e., radiotherapy of brain tumor patients) or unplanned radiation exposure (i.e., accidental radiation exposure) can induce nonspecific damage to neuronal tissues, resulting in chronic or acute radiation syndrome. Although anatomical alterations in the neuronal tissues have been reported at higher doses of gamma radiation, biochemical and molecular perturbations may be evident even at much lower radiation doses. They may manifest in the form of neuronal deficits and cognitive impairment. In the present review, several molecular events and signaling pathways, such as oxidative stress, neuroinflammation, apoptosis, cognition, neuroplasticity, and neurotoxicity induced in neuronal cells upon ionizing radiation exposure, are reviewed. Furthermore, brain-specific radioprotectors and mitigators that protect normal neuronal cells and tissues against ionizing radiation during radiotherapy of cancer patients or nuclear emergencies are also discussed.
Collapse
Affiliation(s)
- Ravi Kumar
- Radiation Biotechnology Department, Institute of Nuclear Medicine and Allied Sciences (INMAS), Defence Research and Development Organization (DRDO), Brig. S.K. Mazumdar Road, Timarpur, Delhi, 110054, India
| | - Pratibha Kumari
- Radiation Biotechnology Department, Institute of Nuclear Medicine and Allied Sciences (INMAS), Defence Research and Development Organization (DRDO), Brig. S.K. Mazumdar Road, Timarpur, Delhi, 110054, India
| | - Raj Kumar
- Radiation Biotechnology Department, Institute of Nuclear Medicine and Allied Sciences (INMAS), Defence Research and Development Organization (DRDO), Brig. S.K. Mazumdar Road, Timarpur, Delhi, 110054, India.
| |
Collapse
|
2
|
Wieg L, Ciola JC, Wasén CC, Gaba F, Colletti BR, Schroeder MK, Hinshaw RG, Ekwudo MN, Holtzman DM, Saito T, Sasaguri H, Saido TC, Cox LM, Lemere CA. Cognitive Effects of Simulated Galactic Cosmic Radiation Are Mediated by ApoE Status, Sex, and Environment in APP Knock-In Mice. Int J Mol Sci 2024; 25:9379. [PMID: 39273325 PMCID: PMC11394682 DOI: 10.3390/ijms25179379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 08/25/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
Cosmic radiation experienced during space travel may increase the risk of cognitive impairment. While simulated galactic cosmic radiation (GCRsim) has led to memory deficits in wildtype (WT) mice, it has not been investigated whether GCRsim in combination with genetic risk factors for Alzheimer's disease (AD) worsens memory further in aging mice. Here, we investigated the central nervous system (CNS) effects of 0 Gy (sham) or 0.75 Gy five-ion GCRsim or 2 Gy gamma radiation (IRR) in 14-month-old female and male APPNL-F/NL-F knock-in (KI) mice bearing humanized ApoE3 or ApoE4 (APP;E3F and APP;E4F). As travel to a specialized facility was required for irradiation, both traveled sham-irradiated C57BL/6J WT and KI mice and non-traveled (NT) KI mice acted as controls for potential effects of travel. Mice underwent four behavioral tests at 20 months of age and were euthanized for pathological and biochemical analyses 1 month later. Fecal samples were collected pre- and post-irradiation at four different time points. GCRsim seemed to impair memory in male APP;E3F mice compared to their sham counterparts. Travel tended to improve cognition in male APP;E3F mice and lowered total Aβ in female and male APP;E3F mice compared to their non-traveled counterparts. Sham-irradiated male APP;E4F mice accumulated more fibrillar amyloid than their APP;E3F counterparts. Radiation exposure had only modest effects on behavior and brain changes, but travel-, sex-, and genotype-specific effects were seen. Irradiated mice had immediate and long-term differences in their gut bacterial composition that correlated to Alzheimer's disease phenotypes.
Collapse
Affiliation(s)
- Laura Wieg
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Boston, MA 02115, USA; (L.W.); (J.C.C.); (C.C.W.); (F.G.); (B.R.C.); (M.K.S.); (R.G.H.); (M.N.E.); (L.M.C.)
| | - Jason C. Ciola
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Boston, MA 02115, USA; (L.W.); (J.C.C.); (C.C.W.); (F.G.); (B.R.C.); (M.K.S.); (R.G.H.); (M.N.E.); (L.M.C.)
| | - Caroline C. Wasén
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Boston, MA 02115, USA; (L.W.); (J.C.C.); (C.C.W.); (F.G.); (B.R.C.); (M.K.S.); (R.G.H.); (M.N.E.); (L.M.C.)
| | - Fidelia Gaba
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Boston, MA 02115, USA; (L.W.); (J.C.C.); (C.C.W.); (F.G.); (B.R.C.); (M.K.S.); (R.G.H.); (M.N.E.); (L.M.C.)
| | - Brianna R. Colletti
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Boston, MA 02115, USA; (L.W.); (J.C.C.); (C.C.W.); (F.G.); (B.R.C.); (M.K.S.); (R.G.H.); (M.N.E.); (L.M.C.)
| | - Maren K. Schroeder
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Boston, MA 02115, USA; (L.W.); (J.C.C.); (C.C.W.); (F.G.); (B.R.C.); (M.K.S.); (R.G.H.); (M.N.E.); (L.M.C.)
| | - Robert G. Hinshaw
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Boston, MA 02115, USA; (L.W.); (J.C.C.); (C.C.W.); (F.G.); (B.R.C.); (M.K.S.); (R.G.H.); (M.N.E.); (L.M.C.)
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Millicent N. Ekwudo
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Boston, MA 02115, USA; (L.W.); (J.C.C.); (C.C.W.); (F.G.); (B.R.C.); (M.K.S.); (R.G.H.); (M.N.E.); (L.M.C.)
| | - David M. Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA;
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Science, Nagoya 467-8601, Aichi, Japan;
| | - Hiroki Sasaguri
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako City 351-0198, Saitama, Japan; (H.S.); (T.C.S.)
| | - Takaomi C. Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako City 351-0198, Saitama, Japan; (H.S.); (T.C.S.)
| | - Laura M. Cox
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Boston, MA 02115, USA; (L.W.); (J.C.C.); (C.C.W.); (F.G.); (B.R.C.); (M.K.S.); (R.G.H.); (M.N.E.); (L.M.C.)
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Cynthia A. Lemere
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Boston, MA 02115, USA; (L.W.); (J.C.C.); (C.C.W.); (F.G.); (B.R.C.); (M.K.S.); (R.G.H.); (M.N.E.); (L.M.C.)
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
3
|
Wang W, Zhao F, Torres S, Harris PL, Wang X, Peng L, Siedlak SL, Zhu X. Space-Like Irradiation Exacerbated Cognitive Deficits and Amyloid Pathology in CRND8 Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2024; 100:S327-S339. [PMID: 39058444 PMCID: PMC11875098 DOI: 10.3233/jad-240570] [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] [Indexed: 07/28/2024]
Abstract
Background Space radiation was linked to neurological damage and behavioral deficits which raised concerns of increased degenerative risk on the brain and development of Alzheimer's disease following space travel. Objective In this study, we investigated the effects of irradiation by 56Fe and 28Si in CRND8 mice, an Alzheimer's disease mouse model. Methods Six-month-old CRND8 mice were exposed to whole body irradiation by 56Fe and 28Si at 0.5 Gy and 2 Gy doses. Behavior tests were administered 1-month to 3-months post-irradiation. Amyloid deposition and other pathological changes were analyzed 3-months and/or 6-months post-irradiation. Results The Novel Object Recognition test showed some decline in 8-month-old mice compared to non-irradiated CRND8 mice. Male mice also showed a loss of freezing behavior in the fear conditioning contextual test following irradiation. Golgi staining revealed a loss of spines in hippocampal neurons after irradiation. Total amyloid immunohistochemistry showed a robust increase in 3-months post-irradiation 56Fe groups which became normalized to non-irradiated group by 6-months post-irradiation. However, 2 Gy 28Si caused a trend towards increased plaque load at 3-months post-irradiation which became significant at 6-months post irradiation only in male CRND8 mice. While 0.5 Gy Fe did not induce obvious changes in the total number of iba-1 positive microglia, more hippocampal microglia were found to express PCNA after 0.5 Gy Fe treatment, suggesting potential involvement of microglial dysfunction. Conclusions Overall, our study provides new evidence of gender-specific and ion-dependent effects of space radiation on cognition and amyloid pathology in AD models.
Collapse
Affiliation(s)
- Wenzhang Wang
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Fanpeng Zhao
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Sandy Torres
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Peggy L.R. Harris
- Department of Genetics, Case Western Reserve University, Cleveland, OH, USA
| | - Xinglong Wang
- Department of Pharmacology & Toxicology, University of Arizona, Tucson, AZ, USA
| | - Lihua Peng
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Sandra L. Siedlak
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Xiongwei Zhu
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
4
|
Pathare NN, Fayet-Moore F, Fogarty JA, Jacka FN, Strandwitz P, Strangman GE, Donoviel DB. Nourishing the brain on deep space missions: nutritional psychiatry in promoting resilience. Front Neural Circuits 2023; 17:1170395. [PMID: 37663891 PMCID: PMC10469890 DOI: 10.3389/fncir.2023.1170395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 07/31/2023] [Indexed: 09/05/2023] Open
Abstract
The grueling psychological demands of a journey into deep space coupled with ever-increasing distances away from home pose a unique problem: how can we best take advantage of the benefits of fresh foods in a place that has none? Here, we consider the biggest challenges associated with our current spaceflight food system, highlight the importance of supporting optimal brain health on missions into deep space, and discuss evidence about food components that impact brain health. We propose a future food system that leverages the gut microbiota that can be individually tailored to best support the brain and mental health of crews on deep space long-duration missions. Working toward this goal, we will also be making investments in sustainable means to nourish the crew that remains here on spaceship Earth.
Collapse
Affiliation(s)
- Nihar N. Pathare
- Center for Space Medicine, Baylor College of Medicine, Houston, TX, United States
| | | | - Jennifer A. Fogarty
- Center for Space Medicine, Baylor College of Medicine, Houston, TX, United States
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
- Translational Research Institute for Space Health (TRISH), Houston, TX, United States
| | - Felice N. Jacka
- Food and Mood Centre, Institute for Mental and Physical Health and Clinical Translation (IMPACT) Strategic Research Centre, Deakin University, Geelong, VIC, Australia
- Department of Psychiatry, The University of Melbourne, Parkville, VIC, Australia
| | | | - Gary E. Strangman
- Neural Systems Group, Division of Health Sciences and Technology, Massachusetts General Hospital, Harvard Medical School and Harvard-MIT, Charlestown, MA, United States
- Department of Psychology, Harvard University, Cambridge, MA, United States
| | - Dorit B. Donoviel
- Center for Space Medicine, Baylor College of Medicine, Houston, TX, United States
- Translational Research Institute for Space Health (TRISH), Houston, TX, United States
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
5
|
Faerman A, Clark JB, Sutton JP. Neuropsychological considerations for long-duration deep spaceflight. Front Physiol 2023; 14:1146096. [PMID: 37275233 PMCID: PMC10235498 DOI: 10.3389/fphys.2023.1146096] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 05/09/2023] [Indexed: 06/07/2023] Open
Abstract
The deep space environment far beyond low-Earth orbit (LEO) introduces multiple and simultaneous risks for the functioning and health of the central nervous system (CNS), which may impair astronauts' performance and wellbeing. As future deep space missions to Mars, moons, or asteroids will also exceed current LEO stay durations and are estimated to require up to 3 years, we review recent evidence with contemporary and historic spaceflight case studies addressing implications for long-duration missions. To highlight the need for specific further investigations, we provide neuropsychological considerations integrating cognitive and motor functions, neuroimaging, neurological biomarkers, behavior changes, and mood and affect to construct a multifactorial profile to explain performance variability, subjective experience, and potential risks. We discuss the importance of adopting a neuropsychological approach to long-duration deep spaceflight (LDDS) missions and draw specific recommendations for future research in space neuropsychology.
Collapse
Affiliation(s)
- Afik Faerman
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, United States
| | - Jonathan B. Clark
- Center for Space Medicine, Baylor College of Medicine, Houston, TX, United States
- Department of Neurology, Baylor College of Medicine, Houston, TX, United States
| | - Jeffrey P. Sutton
- Center for Space Medicine, Baylor College of Medicine, Houston, TX, United States
- Translational Research Institute for Space Health, Baylor College of Medicine, Houston, TX, United States
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
6
|
Bok J, Ha J, Ahn BJ, Jang Y. Disease-Modifying Effects of Non-Invasive Electroceuticals on β-Amyloid Plaques and Tau Tangles for Alzheimer's Disease. Int J Mol Sci 2022; 24:ijms24010679. [PMID: 36614120 PMCID: PMC9821138 DOI: 10.3390/ijms24010679] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/22/2022] [Accepted: 12/27/2022] [Indexed: 01/04/2023] Open
Abstract
Electroceuticals refer to various forms of electronic neurostimulators used for therapy. Interdisciplinary advances in medical engineering and science have led to the development of the electroceutical approach, which involves therapeutic agents that specifically target neural circuits, to realize precision therapy for Alzheimer's disease (AD). To date, extensive studies have attempted to elucidate the disease-modifying effects of electroceuticals on areas in the brain of a patient with AD by the use of various physical stimuli, including electric, magnetic, and electromagnetic waves as well as ultrasound. Herein, we review non-invasive stimulatory systems and their effects on β-amyloid plaques and tau tangles, which are pathological molecular markers of AD. Therefore, this review will aid in better understanding the recent technological developments, applicable methods, and therapeutic effects of electronic stimulatory systems, including transcranial direct current stimulation, 40-Hz gamma oscillations, transcranial magnetic stimulation, electromagnetic field stimulation, infrared light stimulation and ionizing radiation therapy, and focused ultrasound for AD.
Collapse
Affiliation(s)
- Junsoo Bok
- Department of Medical and Digital Engineering, College of Engineering, Hanyang University, Seoul 04736, Republic of Korea
| | - Juchan Ha
- Department of Biomedical Engineering, College of Engineering, Hanyang University, Seoul 04736, Republic of Korea
| | - Bum Ju Ahn
- Department of Pharmacology, College of Medicine, Hanyang University, Seoul 04736, Republic of Korea
| | - Yongwoo Jang
- Department of Medical and Digital Engineering, College of Engineering, Hanyang University, Seoul 04736, Republic of Korea
- Department of Pharmacology, College of Medicine, Hanyang University, Seoul 04736, Republic of Korea
- Correspondence: ; Tel.: +82-2-2220-0655
| |
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: 0.7] [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
|
Desai RI, Limoli CL, Stark CEL, Stark SM. Impact of spaceflight stressors on behavior and cognition: A molecular, neurochemical, and neurobiological perspective. Neurosci Biobehav Rev 2022; 138:104676. [PMID: 35461987 DOI: 10.1016/j.neubiorev.2022.104676] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 03/15/2022] [Accepted: 04/18/2022] [Indexed: 11/19/2022]
Abstract
The response of the human body to multiple spaceflight stressors is complex, but mounting evidence implicate risks to CNS functionality as significant, able to threaten metrics of mission success and longer-term behavioral and neurocognitive health. Prolonged exposure to microgravity, sleep disruption, social isolation, fluid shifts, and ionizing radiation have been shown to disrupt mechanisms of homeostasis and neurobiological well-being. The overarching goal of this review is to document the existing evidence of how the major spaceflight stressors, including radiation, microgravity, isolation/confinement, and sleep deprivation, alone or in combination alter molecular, neurochemical, neurobiological, and plasma metabolite/lipid signatures that may be linked to operationally-relevant behavioral and cognitive performance. While certain brain region-specific and/or systemic alterations titrated in part with neurobiological outcome, variations across model systems, study design, and the conspicuous absence of targeted studies implementing combinations of spaceflight stressors, confounded the identification of specific signatures having direct relevance to human activities in space. Summaries are provided for formulating new research directives and more predictive readouts of portending change in neurobiological function.
Collapse
Affiliation(s)
- Rajeev I Desai
- Harvard Medical School, McLean Hospital, Behavioral Biology Program, Belmont, MA 02478, USA.
| | - Charles L Limoli
- Department of Radiation Oncology, University of California Irvine, Medical Sciences I, B146B, Irvine, CA 92697, USA
| | - Craig E L Stark
- Department of Neurobiology of Behavior, University of California Irvine, 1400 Biological Sciences III, Irvine, CA 92697, USA
| | - Shauna M Stark
- Department of Neurobiology of Behavior, University of California Irvine, 1400 Biological Sciences III, Irvine, CA 92697, USA
| |
Collapse
|
9
|
Kiffer FC, Luitel K, Tran FH, Patel RA, Guzman CS, Soler I, Xiao R, Shay JW, Yun S, Eisch AJ. Effects of a 33-ion sequential beam galactic cosmic ray analog on male mouse behavior and evaluation of CDDO-EA as a radiation countermeasure. Behav Brain Res 2022; 419:113677. [PMID: 34818568 PMCID: PMC9755463 DOI: 10.1016/j.bbr.2021.113677] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/28/2021] [Accepted: 11/16/2021] [Indexed: 12/21/2022]
Abstract
In long-term spaceflight, astronauts will face unique cognitive loads and social challenges which will be complicated by communication delays with Earth. It is important to understand the central nervous system (CNS) effects of deep spaceflight and the associated unavoidable exposure to galactic cosmic radiation (GCR). Rodent studies show single- or simple-particle combination exposure alters CNS endpoints, including hippocampal-dependent behavior. An even better Earth-based simulation of GCR is now available, consisting of a 33-beam (33-GCR) exposure. However, the effect of whole-body 33-GCR exposure on rodent behavior is unknown, and no 33-GCR CNS countermeasures have been tested. Here astronaut-age-equivalent (6mo-old) C57BL/6J male mice were exposed to 33-GCR (75cGy, a Mars mission dose). Pre-/during/post-Sham or 33-GCR exposure, mice received a diet containing a 'vehicle' formulation alone or with the antioxidant/anti-inflammatory compound CDDO-EA as a potential countermeasure. Behavioral testing beginning 4mo post-irradiation suggested radiation and diet did not affect measures of exploration/anxiety-like behaviors (open field, elevated plus maze) or recognition of a novel object. However, in 3-Chamber Social Interaction (3-CSI), CDDO-EA/33-GCR mice failed to spend more time exploring a holder containing a novel mouse vs. a novel object (empty holder), suggesting sociability deficits. Also, Vehicle/33-GCR and CDDO-EA/Sham mice failed to discriminate between a novel stranger vs. familiarized stranger mouse, suggesting blunted preference for social novelty. CDDO-EA given pre-/during/post-irradiation did not attenuate the 33-GCR-induced blunting of preference for social novelty. Future elucidation of the mechanisms underlying 33-GCR-induced blunting of preference for social novelty will improve risk analysis for astronauts which may in-turn improve countermeasures.
Collapse
Affiliation(s)
- Frederico C Kiffer
- Department of Anesthesiology and Critical Care Medicine, The Children’s Hospital of Philadelphia (CHOP) Research Institute, Philadelphia, PA, USA, 19104
| | - Krishna Luitel
- Department of Cell Biology, University of Texas Southwestern (UTSW) Medical Center, Dallas, TX, USA, 75390
| | - Fionya H Tran
- Department of Anesthesiology and Critical Care Medicine, The Children’s Hospital of Philadelphia (CHOP) Research Institute, Philadelphia, PA, USA, 19104
| | - Riya A Patel
- Department of Anesthesiology and Critical Care Medicine, The Children’s Hospital of Philadelphia (CHOP) Research Institute, Philadelphia, PA, USA, 19104
| | - Catalina S Guzman
- Department of Anesthesiology and Critical Care Medicine, The Children’s Hospital of Philadelphia (CHOP) Research Institute, Philadelphia, PA, USA, 19104
| | - Ivan Soler
- Department of Anesthesiology and Critical Care Medicine, The Children’s Hospital of Philadelphia (CHOP) Research Institute, Philadelphia, PA, USA, 19104
| | - Rui Xiao
- Department of Pediatrics Division of Biostatistics, CHOP Research Institute, Philadelphia, PA, USA, 19104,Department of Biostatistics, Epidemiology & Informatics, University of Pennsylvania, Philadelphia, PA, USA, 19104
| | - Jerry W Shay
- Department of Cell Biology, University of Texas Southwestern (UTSW) Medical Center, Dallas, TX, USA, 75390
| | - Sanghee Yun
- Department of Anesthesiology and Critical Care Medicine, The Children’s Hospital of Philadelphia (CHOP) Research Institute, Philadelphia, PA, USA, 19104,Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA, 19104
| | - Amelia J Eisch
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia (CHOP) Research Institute, Philadelphia, PA 19104, USA; Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
10
|
Lalkovicova M. Neuroprotective agents effective against radiation damage of central nervous system. Neural Regen Res 2022; 17:1885-1892. [PMID: 35142663 PMCID: PMC8848589 DOI: 10.4103/1673-5374.335137] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Ionizing radiation caused by medical treatments, nuclear events or even space flights can irreversibly damage structure and function of brain cells. That can result in serious brain damage, with memory and behavior disorders, or even fatal oncologic or neurodegenerative illnesses. Currently used treatments and drugs are mostly targeting biochemical processes of cell apoptosis, radiation toxicity, neuroinflammation, and conditions such as cognitive-behavioral disturbances or others that result from the radiation insult. With most drugs, the side effects and potential toxicity are also to be considered. Therefore, many agents have not been approved for clinical use yet. In this review, we focus on the latest and most effective agents that have been used in animal and also in the human research, and clinical treatments. They could have the potential therapeutical use in cases of radiation damage of central nervous system, and also in prevention considering their radioprotecting effect of nervous tissue.
Collapse
Affiliation(s)
- Mária Lalkovicova
- Laboratory of Radiation Biology, Joint Institute for Nuclear Research, Dubna, Russia; Slovak Academy of Sciences, Institute of Experimental Physics, Košice, Slovakia
| |
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: 2.3] [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
|
Mhatre SD, Iyer J, Puukila S, Paul AM, Tahimic CGT, Rubinstein L, Lowe M, Alwood JS, Sowa MB, Bhattacharya S, Globus RK, Ronca AE. Neuro-consequences of the spaceflight environment. Neurosci Biobehav Rev 2021; 132:908-935. [PMID: 34767877 DOI: 10.1016/j.neubiorev.2021.09.055] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 08/03/2021] [Accepted: 09/28/2021] [Indexed: 12/17/2022]
Abstract
As human space exploration advances to establish a permanent presence beyond the Low Earth Orbit (LEO) with NASA's Artemis mission, researchers are striving to understand and address the health challenges of living and working in the spaceflight environment. Exposure to ionizing radiation, microgravity, isolation and other spaceflight hazards pose significant risks to astronauts. Determining neurobiological and neurobehavioral responses, understanding physiological responses under Central Nervous System (CNS) control, and identifying putative mechanisms to inform countermeasure development are critically important to ensuring brain and behavioral health of crew on long duration missions. Here we provide a detailed and comprehensive review of the effects of spaceflight and of ground-based spaceflight analogs, including simulated weightlessness, social isolation, and ionizing radiation on humans and animals. Further, we discuss dietary and non-dietary countermeasures including artificial gravity and antioxidants, among others. Significant future work is needed to ensure that neural, sensorimotor, cognitive and other physiological functions are maintained during extended deep space missions to avoid potentially catastrophic health and safety outcomes.
Collapse
Affiliation(s)
- Siddhita D Mhatre
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA; KBR, Houston, TX, 77002, USA; COSMIAC Research Center, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Janani Iyer
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA; Universities Space Research Association, Columbia, MD, 21046, USA
| | - Stephanie Puukila
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA; Universities Space Research Association, Columbia, MD, 21046, USA; Flinders University, Adelaide, Australia
| | - Amber M Paul
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA; Universities Space Research Association, Columbia, MD, 21046, USA
| | - Candice G T Tahimic
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA; KBR, Houston, TX, 77002, USA; Department of Biology, University of North Florida, Jacksonville, FL, 32224, USA
| | - Linda Rubinstein
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA; Universities Space Research Association, Columbia, MD, 21046, USA
| | - Moniece Lowe
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA; Blue Marble Space Institute of Science, Seattle, WA, 98154, USA
| | - Joshua S Alwood
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA
| | - Marianne B Sowa
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA
| | - Sharmila Bhattacharya
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA
| | - Ruth K Globus
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA
| | - April E Ronca
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA; Wake Forest Medical School, Winston-Salem, NC, 27101, USA.
| |
Collapse
|
13
|
Dissmore T, DeMarco AG, Jayatilake M, Girgis M, Bansal S, Li Y, Mehta K, Sridharan V, Gill K, Bansal S, Tyburski JB, Cheema AK. Longitudinal metabolic alterations in plasma of rats exposed to low doses of high linear energy transfer radiation. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART C, TOXICOLOGY AND CARCINOGENESIS 2021; 39:219-233. [PMID: 33902389 PMCID: PMC9896584 DOI: 10.1080/26896583.2020.1865027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Astronauts embarking on deep space missions are at high risk of long-term exposure to low doses of high linear energy transfer (LET) radiation, which can contribute to the development of cancer and multiple degenerative diseases. However, long term effects of exposure to low doses of high LET radiation in plasma metabolite profiles have not been elucidated. We utilized an untargeted metabolomics and lipidomics approach to analyze plasma obtained from adult male Long Evans rats to determine the longitudinal effects of low-dose proton and low-dose oxygen ion whole-body irradiation on metabolic pathways. Our findings reveal that radiation exposure induced modest changes in the metabolic profiles in plasma, 7 months after exposure. Furthermore, we identified some common metabolite dysregulations between protons and oxygen ions, which may indicate a similar mechanism of action for both radiation types.
Collapse
Affiliation(s)
- Tixieanna Dissmore
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Medical Center, Washington, DC, USA
| | - Andrew G DeMarco
- Department of Biochemistry and Molecular Biology, Georgetown University Medical Center, Washington, DC, USA
| | - Meth Jayatilake
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Medical Center, Washington, DC, USA
| | - Michael Girgis
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Medical Center, Washington, DC, USA
| | - Shivani Bansal
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Medical Center, Washington, DC, USA
| | - Yaoxiang Li
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Medical Center, Washington, DC, USA
| | - Khyati Mehta
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Medical Center, Washington, DC, USA
| | - Vijayalakshmi Sridharan
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Kirandeep Gill
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Medical Center, Washington, DC, USA
| | - Sunil Bansal
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Medical Center, Washington, DC, USA
| | | | - Amrita K Cheema
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Medical Center, Washington, DC, USA
- Department of Biochemistry and Molecular Biology, Georgetown University Medical Center, Washington, DC, USA
| |
Collapse
|
14
|
Romanella SM, Sprugnoli G, Ruffini G, Seyedmadani K, Rossi S, Santarnecchi E. Noninvasive Brain Stimulation & Space Exploration: Opportunities and Challenges. Neurosci Biobehav Rev 2020; 119:294-319. [PMID: 32937115 PMCID: PMC8361862 DOI: 10.1016/j.neubiorev.2020.09.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 05/22/2020] [Accepted: 09/03/2020] [Indexed: 01/11/2023]
Abstract
As NASA prepares for longer space missions aiming for the Moon and Mars, astronauts' health and performance are becoming a central concern due to the threats associated with galactic cosmic radiation, unnatural gravity fields, and life in extreme environments. In space, the human brain undergoes functional and structural changes related to fluid shift and changes in intracranial pressure. Behavioral abnormalities, such as cognitive deficits, sleep disruption, and visuomotor difficulties, as well as psychological effects, are also an issue. We discuss opportunities and challenges of noninvasive brain stimulation (NiBS) methods - including transcranial magnetic stimulation (TMS) and transcranial electrical stimulation (tES) - to support space exploration in several ways. NiBS includes safe and portable techniques already applied in a wide range of cognitive and motor domains, as well as therapeutically. NiBS could be used to enhance in-flight performance, supporting astronauts during pre-flight Earth-based training, as well as to identify biomarkers of post-flight brain changes for optimization of rehabilitation/compensatory strategies. We review these NiBS techniques and their effects on brain physiology, psychology, and cognition.
Collapse
Affiliation(s)
- S M Romanella
- Siena Brain Investigation & Neuromodulation Lab (Si-BIN Lab), Department of Medicine, Surgery and Neuroscience, Neurology and Clinical Neurophysiology Section, University of Siena, Italy
| | - G Sprugnoli
- Berenson-Allen Center for Noninvasive Brain Stimulation, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Radiology Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - G Ruffini
- Neuroelectrics Corporation, Cambridge, MA, USA
| | - K Seyedmadani
- University Space Research Association NASA Johnson Space Center, Houston, TX, USA; Ann and H.J. Smead Aerospace Engineering Sciences, University of Colorado, Boulder, CO, USA
| | - S Rossi
- Siena Brain Investigation & Neuromodulation Lab (Si-BIN Lab), Department of Medicine, Surgery and Neuroscience, Neurology and Clinical Neurophysiology Section, University of Siena, Italy; Human Physiology Section, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - E Santarnecchi
- Siena Brain Investigation & Neuromodulation Lab (Si-BIN Lab), Department of Medicine, Surgery and Neuroscience, Neurology and Clinical Neurophysiology Section, University of Siena, Italy; Berenson-Allen Center for Noninvasive Brain Stimulation, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
15
|
Cahoon DS, Shukitt-Hale B, Bielinski DF, Hawkins EM, Cacioppo AM, Rabin BM. Effects of partial- or whole-body exposures to 56Fe particles on brain function and cognitive performance in rats. LIFE SCIENCES IN SPACE RESEARCH 2020; 27:56-63. [PMID: 34756230 DOI: 10.1016/j.lssr.2020.07.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/01/2020] [Accepted: 07/22/2020] [Indexed: 05/03/2023]
Abstract
On exploratory class missions, such as a mission to Mars, astronauts will be exposed to particles of high energy and charge (HZE particles). Exposure to HZE particles produces changes in neuronal function and can disrupt cognitive performance. Cells throughout the entire body, not just the brain, will be impacted by these particles. To determine the possible effects that irradiation of the body might have on neuronal function and cognitive performance, rats were given head-only, body-only or whole-body exposures to 56Fe particles. Cognitive performance (novel object recognition, operant responding) was tested in one set of animals; changes in brain function (oxidative stress, neuroinflammation) was tested in a second set of rats. The results indicated that there were no consistent differences in either behavioral or neurochemical endpoints as a function of the location of the irradiation. These results suggest that radiation to the body can impact the brain, therefore it may be necessary to re-evaluate the estimates of the risk of HZE particle-induced changes in neuronal function and cognitive performance.
Collapse
Affiliation(s)
- Danielle S Cahoon
- USDA-ARS, Human Nutrition Research Center on Aging at Tufts Univ., Boston, MA 02111, USA
| | - Barbara Shukitt-Hale
- USDA-ARS, Human Nutrition Research Center on Aging at Tufts Univ., Boston, MA 02111, USA
| | - Donna F Bielinski
- USDA-ARS, Human Nutrition Research Center on Aging at Tufts Univ., Boston, MA 02111, USA
| | | | | | | |
Collapse
|
16
|
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: 79] [Impact Index Per Article: 15.8] [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
|
17
|
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: 46] [Impact Index Per Article: 7.7] [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
|
18
|
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: 55] [Impact Index Per Article: 9.2] [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
|
19
|
Batmunkh M, Aksenova SV, Bayarchimeg L, Bugay AN, Lkhagva O. Optimized neuron models for estimation of charged particle energy deposition in hippocampus. Phys Med 2019; 57:88-94. [PMID: 30738537 DOI: 10.1016/j.ejmp.2019.01.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 12/29/2018] [Accepted: 01/01/2019] [Indexed: 01/31/2023] Open
Abstract
The study of evaluating radiation risk on the central nervous system induced by space-born charged particles is very complex and challenging task in space radiobiology and radiation protection. To overcome computational difficulties in this field, we developed simplified neuron models with properties equivalent to realistic neuron morphology. Three-dimensional structure and parameters of simplified and complex neuron models with realistic morphology were obtained from the experimental data. The models implement uniform random distribution of spines along the dendritic branches in typical hippocampal neurons. Both types of models were implemented and tested using Geant4 Monte Carlo radiation transport code. Track structure simulations were performed for ion beams with typical fluxes of galactic cosmic rays expected for long-term interplanetary missions. The distribution of energy deposition events and percentage of irradiated volumes were obtained to be similar in both simplified and realistic models of pyramidal and granule cells of the rat hippocampus following irradiation. Significant increase of computational efficiency for detailed microdosimetry simulations of hippocampus using simplified neuron models was achieved. Using designed neuron models we have constructed 3D model of the rat hippocampus, including pyramidal cells, mature and immature granular cells, mossy cells, and neural stem cells. Computed energy deposition in irradiated hippocampal neurons following a track of iron ion suggests that most of energy is accumulated by dense population of granular cells in the dentate gyrus. Proposed approach could serve as a complementary computation technique for studying radiation-induced effects in large scale brain networks.
Collapse
Affiliation(s)
- Munkhbaatar Batmunkh
- Laboratory of Radiation Biology, Joint Institute for Nuclear Research, Dubna 141980, Russia.
| | - Svetlana V Aksenova
- Laboratory of Radiation Biology, Joint Institute for Nuclear Research, Dubna 141980, Russia.
| | - Lkhagvaa Bayarchimeg
- Laboratory of Radiation Biology, Joint Institute for Nuclear Research, Dubna 141980, Russia.
| | - Aleksandr N Bugay
- Laboratory of Radiation Biology, Joint Institute for Nuclear Research, Dubna 141980, Russia.
| | - Oidov Lkhagva
- Division of Natural Sciences, National University of Mongolia, Ulaanbaatar 210646, Mongolia.
| |
Collapse
|
20
|
Batmunkh M, Bayarchimeg L, Bugay AN, Lkhagva O. Monte Carlo track structure simulation in studies of biological effects induced by accelerated charged particles in the central nervous system. EPJ WEB OF CONFERENCES 2019. [DOI: 10.1051/epjconf/201920404008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Simulating the biological damage induced by charged particles trajectories (tracks) in the central nervous system (CNS) at different levels of its organization (molecular, cellular, and tissue) is a challenge of modern radiobiology studies. According to the recent experimental studies at particle accelerators, the most radiation-sensitive area of the CNS is the hippocampus. In this regards, the development of measurement-based Monte Carlo simulation of radiation-induced alterations in the hippocampus is of great interest to understand the radiobiological effects on the CNS. The present work investigates the influence of charged particles on the hippocampal cells of the rat brain using the Geant4 Monte Carlo radiation transport code. The applied computer simulation provides a method to simulate physics processes and chemical reactions in the developed model of the rat hippocampus, which contains different types of neural cells - pyramidal cells, mature and immature granular cells, mossy cells, and neural stem cells. The distribution of stochastic energy depositions has been obtained and analyzed in critical structures of the hippocampal neurons after irradiation with 600 MeV/u iron particles. The computed energy deposition in irradiated hippocampal neurons following a track of iron ion suggests that most of the energy is accumulated by granular cells. The obtained quantities at the level of molecular targets also assume that NMDA and GABA receptors belong to the most probable targets in the irradiated neural cells.
Collapse
|
21
|
Heselich A, Frieß JL, Ritter S, Benz NP, Layer PG, Thielemann C. High LET radiation shows no major cellular and functional effects on primary cardiomyocytes in vitro. LIFE SCIENCES IN SPACE RESEARCH 2018; 16:93-100. [PMID: 29475525 DOI: 10.1016/j.lssr.2018.01.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 12/18/2017] [Accepted: 01/10/2018] [Indexed: 06/08/2023]
Abstract
It is well known that ionizing radiation causes adverse effects on various mammalian tissues. However, there is little information on the biological effects of heavy ion radiation on the heart. In order to fill this gap, we systematically examined DNA-damage induction and repair, as well as proliferation and apoptosis in avian cardiomyocyte cultures irradiated with heavy ions such as titanium and iron, relevant for manned space-flight, and carbon ions, as used for radiotherapy. Further, and to our knowledge for the first time, we analyzed the effect of heavy ion radiation on the electrophysiology of primary cardiomyocytes derived from chicken embryos using the non-invasive microelectrode array (MEA) technology. As electrophysiological endpoints beat rate and field action potential duration were analyzed. The cultures clearly exhibited the capacity to repair induced DNA damage almost completely within 24 h, even at doses of 7 Gy, and almost completely recovered from radiation-induced changes in proliferative behavior. Interestingly, no significant effects on apoptosis could be detected. Especially the functionality of primary cardiac cells exhibited a surprisingly high robustness against heavy ion radiation, even at doses of up to 7 Gy. In contrast to our previous study with X-rays the beat rate remained more or less unaffected after heavy ion radiation, independently of beam quality. The only change we could observe was an increase of the field action potential duration of up to 30% after titanium irradiation, diminishing within the following three days. This potentially pathological observation may be an indication that heavy ion irradiation at high doses could bear a long-term risk for cardiovascular disease induction.
Collapse
Affiliation(s)
- Anja Heselich
- University for Applied Sciences Aschaffenburg, biomems lab, Würzburger Straße 45, Aschaffenburg 63743, Germany; Technische Universität Darmstadt, Developmental Biology and Neurogenetics, Schnittspahnstraße 13, Darmstadt 64287, Germany; GSI Helmholtz Centre for Heavy Ion Research (GSI), Biophysics Department, Planckstraße 1, Darmstadt 64291, Germany
| | - Johannes L Frieß
- University for Applied Sciences Aschaffenburg, biomems lab, Würzburger Straße 45, Aschaffenburg 63743, Germany
| | - Sylvia Ritter
- GSI Helmholtz Centre for Heavy Ion Research (GSI), Biophysics Department, Planckstraße 1, Darmstadt 64291, Germany
| | - Naja P Benz
- Technische Universität Darmstadt, Developmental Biology and Neurogenetics, Schnittspahnstraße 13, Darmstadt 64287, Germany
| | - Paul G Layer
- Technische Universität Darmstadt, Developmental Biology and Neurogenetics, Schnittspahnstraße 13, Darmstadt 64287, Germany
| | - Christiane Thielemann
- University for Applied Sciences Aschaffenburg, biomems lab, Würzburger Straße 45, Aschaffenburg 63743, Germany.
| |
Collapse
|
22
|
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: 32] [Impact Index Per Article: 4.0] [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
|
23
|
Tang FR, Loke WK, Wong P, Khoo BC. Radioprotective effect of ursolic acid in radiation-induced impairment of neurogenesis, learning and memory in adolescent BALB/c mouse. Physiol Behav 2017; 175:37-46. [PMID: 28341234 DOI: 10.1016/j.physbeh.2017.03.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 03/13/2017] [Accepted: 03/20/2017] [Indexed: 10/19/2022]
Abstract
The effect of acute irradiation with 5Gy or fractionated exposure with 0.5Gy continuously for 10days (a total dose of 5Gy) was evaluated in an immature BALB/c mouse model. Radioprotective effect of ursolic acid (at 25mg/kg/daily administered 1h after acute or each of fractionated irradiations, and continuously for 30days) was also investigated. We found that both acute and fractionated irradiation at a total dose of 5Gy did not induce any mortality within 30days after exposure to postnatal day 26 (P26) BALB/c mice, but reduced animal weigh gain in the first few weeks. At 90days after irradiation, the weight of animals with acute irradiation was still significantly lower than the control group; no significant difference though was observed for those fractionatedly exposed mice compared to the control group. Behavioral tests indicated that acute irradiation at 5Gy induced deficits in learning and memory in the contextual fear conditioning test. The memory for novel object recognition was also impaired. Similar changes were not observed in mice with fractionated irradiation. Immunohistochemical study demonstrated clearly that acute and fractionated irradiations induced impairment of neurogenesis in the subgranular zone (SGZ) of the dentate gyrus although fractionated exposure induced much lesser loss of newly generated neurons. Ursolic acid administered at 25mg/kg/daily for 30days after irradiation greatly improved acute irradiation-induced deficits in contextual learning and memory and in novel object recognition memory although it exacerbated radiation-induced reduction of neurogenesis in SGZ.
Collapse
Affiliation(s)
- Feng Ru Tang
- Radiation Physiology Laboratory, Singapore Nuclear Research and Safety initiative, National University of Singapore, 1 CREATE Way #04-01, CREATE Tower, 138602, Singapore.
| | - Weng Keong Loke
- Defence Medical and Environmental Research Institute, DSO National Laboratories, 11 Stockport Road, 11760, Singapore
| | - Peiyan Wong
- Neuroscience Phenotyping Core, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117456, Singapore
| | - Boo Cheong Khoo
- Temasek Laboratories, National University of Singapore, 5A, Engineering Drive 1, 117411, Singapore
| |
Collapse
|
24
|
Tang FR, Loke WK, Khoo BC. Postnatal irradiation-induced hippocampal neuropathology, cognitive impairment and aging. Brain Dev 2017; 39:277-293. [PMID: 27876394 DOI: 10.1016/j.braindev.2016.11.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 11/04/2016] [Accepted: 11/04/2016] [Indexed: 12/26/2022]
Abstract
Irradiation of the brain in early human life may set abnormal developmental events into motion that last a lifetime, leading to a poor quality of life for affected individuals. While the effect of irradiation at different early developmental stages on the late human life has not been investigated systematically, animal experimental studies suggest that acute postnatal irradiation with ⩾0.1Gy may significantly reduce neurogenesis in the dentate gyrus and endotheliogenesis in cerebral vessels and induce cognitive impairment and aging. Fractionated irradiation also reduces neurogenesis. Furthermore, irradiation induces hippocampal neuronal loss in CA1 and CA3 areas, neuroinflammation and reduces gliogenesis. The hippocampal neurovascular niche and the total number of microvessels are also changed after radiation exposures. Each or combination of these pathological changes may cause cognitive impairment and aging. Interestingly, acute irradiation of aged brain with a certain amount of radiation has also been reported to induce brain hormesis or neurogenesis. At molecular levels, inflammatory cytokines, chemokines, neural growth factors, neurotransmitters, their receptors and signal transduction systems, reactive oxygen species are involved in radiation-induced adverse effect on brain development and functions. Further study at different omics levels after low dose/dose rate irradiation may not only unravel the mechanisms of radiation-induced adverse brain effect or hormesis, but also provide clues for detection or diagnosis of radiation exposure and for therapeutic approaches to effectively prevent radiation-induced cognitive impairment and aging. Investigation focusing on radiation-induced changes of critical brain development events may reveal many previously unknown adverse effects.
Collapse
Affiliation(s)
- Feng Ru Tang
- Singapore Nuclear Research and Safety Initiative, National University of Singapore, Singapore 138602, Singapore.
| | - Weng Keong Loke
- Defence Medical and Environmental Research Institute, DSO National Laboratories, 11 Stockport Road, Singapore 11760, Singapore
| | - Boo Cheong Khoo
- Temasek Laboratories, National University of Singapore, 5A, Engineering Drive 1, Singapore 117411, Singapore
| |
Collapse
|
25
|
Raber J, Marzulla T, Kronenberg A, Turker MS. (16)Oxygen irradiation enhances cued fear memory in B6D2F1 mice. LIFE SCIENCES IN SPACE RESEARCH 2015; 7:61-65. [PMID: 26553639 DOI: 10.1016/j.lssr.2015.10.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 10/07/2015] [Accepted: 10/08/2015] [Indexed: 06/05/2023]
Abstract
The space radiation environment includes energetic charged particles that may impact cognitive performance. We assessed the effects of (16)O ion irradiation on cognitive performance of C57BL/6J × DBA/2J F1 (B6D2F1) mice at OHSU (Portland, OR) one month following irradiation at Brookhaven National Laboratory (BNL, Upton, NY). Hippocampus-dependent contextual fear memory and hippocampus-independent cued fear memory of B6D2F1 mice were tested. (16)O ion exposure enhanced cued fear memory. This effect showed a bell-shaped dose response curve. Cued fear memory was significantly stronger in mice irradiated with (16)O ions at a dose of 0.4 or 0.8 Gy than in sham-irradiated mice or following irradiation at 1.6 Gy. In contrast to cued fear memory, contextual fear memory was not affected following (16)O ion irradiation at the doses used in this study. These data indicate that the amygdala might be particularly susceptible to effects of (16)O ion exposure.
Collapse
Affiliation(s)
- Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR 97239, USA; Departments of Neurology and Radiation Medicine, Division of Neuroscience ONPRC, Oregon Health and Science University, Portland, OR 97239, USA.
| | - Tessa Marzulla
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR 97239, USA
| | - Amy Kronenberg
- Department of Cell and Molecular Biology, Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Mitchell S Turker
- Oregon Institute of Occupational Health Sciences and Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR 97239, USA
| |
Collapse
|
26
|
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: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
27
|
Rudobeck E, Nelson GA, Sokolova IV, Vlkolinský R. 28Silicon Radiation Impairs Neuronal Output in CA1 Neurons of Mouse Ventral Hippocampus without Altering Dendritic Excitability. Radiat Res 2014; 181:407-15. [DOI: 10.1667/rr13484.1] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
28
|
Therapeutic and space radiation exposure of mouse brain causes impaired DNA repair response and premature senescence by chronic oxidant production. Aging (Albany NY) 2014; 5:607-22. [PMID: 23928451 PMCID: PMC3796214 DOI: 10.18632/aging.100587] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Despite recent epidemiological evidences linking radiation exposure and a number of human ailments including cancer, mechanistic understanding of how radiation inflicts long-term changes in cerebral cortex, which regulates important neuronal functions, remains obscure. The current study dissects molecular events relevant to pathology in cerebral cortex of 6 to 8 weeks old female C57BL/6J mice two and twelve months after exposure to a γ radiation dose (2 Gy) commonly employed in fractionated radiotherapy. For a comparative study, effects of 1.6 Gy heavy ion 56Fe radiation on cerebral cortex were also investigated, which has implications for space exploration. Radiation exposure was associated with increased chronic oxidative stress, oxidative DNA damage, lipid peroxidation, and apoptosis. These results when considered with decreased cortical thickness, activation of cell-cycle arrest pathway, and inhibition of DNA double strand break repair factors led us to conclude to our knowledge for the first time that radiation caused aging-like pathology in cerebral cortical cells and changes after heavy ion radiation were more pronounced than γ radiation.
Collapse
|
29
|
Cherry JD, Liu B, Frost JL, Lemere CA, Williams JP, Olschowka JA, O’Banion MK. Galactic cosmic radiation leads to cognitive impairment and increased aβ plaque accumulation in a mouse model of Alzheimer's disease. PLoS One 2012; 7:e53275. [PMID: 23300905 PMCID: PMC3534034 DOI: 10.1371/journal.pone.0053275] [Citation(s) in RCA: 142] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2012] [Accepted: 11/27/2012] [Indexed: 01/11/2023] Open
Abstract
Galactic Cosmic Radiation consisting of high-energy, high-charged (HZE) particles poses a significant threat to future astronauts in deep space. Aside from cancer, concerns have been raised about late degenerative risks, including effects on the brain. In this study we examined the effects of (56)Fe particle irradiation in an APP/PS1 mouse model of Alzheimer's disease (AD). We demonstrated 6 months after exposure to 10 and 100 cGy (56)Fe radiation at 1 GeV/µ, that APP/PS1 mice show decreased cognitive abilities measured by contextual fear conditioning and novel object recognition tests. Furthermore, in male mice we saw acceleration of Aβ plaque pathology using Congo red and 6E10 staining, which was further confirmed by ELISA measures of Aβ isoforms. Increases were not due to higher levels of amyloid precursor protein (APP) or increased cleavage as measured by levels of the β C-terminal fragment of APP. Additionally, we saw no change in microglial activation levels judging by CD68 and Iba-1 immunoreactivities in and around Aβ plaques or insulin degrading enzyme, which has been shown to degrade Aβ. However, immunohistochemical analysis of ICAM-1 showed evidence of endothelial activation after 100 cGy irradiation in male mice, suggesting possible alterations in Aβ trafficking through the blood brain barrier as a possible cause of plaque increase. Overall, our results show for the first time that HZE particle radiation can increase Aβ plaque pathology in an APP/PS1 mouse model of AD.
Collapse
Affiliation(s)
- Jonathan D. Cherry
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Bin Liu
- Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jeffrey L. Frost
- Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Cynthia A. Lemere
- Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jacqueline P. Williams
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - John A. Olschowka
- Department of Neurobiology & Anatomy, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - M. Kerry O’Banion
- Department of Neurobiology & Anatomy, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| |
Collapse
|
30
|
Abstract
The complex charged particle environments in space pose considerable challenges with regard to potential health consequences that can impact mission design and crew selection. The lack of knowledge of the biological effects of different ions in isolation and in combination is a particular concern because the risk uncertainties are very high for both cancer and non-cancer late effects. Reducing the uncertainties is of high priority. Two principal components of space radiation each raise different concerns. Solar particle events (SPE) occur sporadically and are comprised primarily of low- to moderate-energy protons. Galactic cosmic radiation (GCR) is isotropic and relatively invariant in dose rate. GCR is also dominated by protons, but the energy range is wider than in SPE. In addition, the contribution of other light and heavy ions to the health risks from GCR must be addressed. This paper will introduce the principal issues under consideration for space radiation protection.
Collapse
Affiliation(s)
- Amy Kronenberg
- Life Sciences Division, Lawrence Berkeley National Laboratory, 1 Cyclotron Road, Berkeley, CA 94720, USA.
| | | |
Collapse
|
31
|
Begum N, Wang B, Mori M, Vares G. Does ionizing radiation influence Alzheimer's disease risk? JOURNAL OF RADIATION RESEARCH 2012; 53:815-22. [PMID: 22872779 PMCID: PMC3483841 DOI: 10.1093/jrr/rrs036] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Alzheimer's disease (AD) is a human neurodegenerative disease, and its global prevalence is predicted to increase dramatically in the following decades. There is mounting evidence describing the effects of ionizing radiation (IR) on the brain, suggesting that exposure to IR might ultimately favor the development of AD. Therefore better understanding the possible connections between exposure to IR and AD pathogenesis is of utmost importance. In this review, recent developments in the research on the biological and cognitive effects of IR in the brain will be explored. Because AD is largely an age-related pathology, the effects of IR on ageing will be investigated.
Collapse
Affiliation(s)
- Nasrin Begum
- Center for Nuclear Medicine and Ultrasound, Rajshahi Medical College Hospital Campus, GPO Box No. 35, Rajshahi, Bangladesh
- National Institute of Radiological Sciences, Anagawa 4-9-1, Inage-ku, Chiba 263-8555, Japan
| | - Bing Wang
- National Institute of Radiological Sciences, Anagawa 4-9-1, Inage-ku, Chiba 263-8555, Japan
| | - Masahiko Mori
- National Institute of Radiological Sciences, Anagawa 4-9-1, Inage-ku, Chiba 263-8555, Japan
| | - Guillaume Vares
- National Institute of Radiological Sciences, Anagawa 4-9-1, Inage-ku, Chiba 263-8555, Japan
- Corresponding author. Tel: +81-(0)43-206-4730;
| |
Collapse
|
32
|
Britten RA, Davis LK, Johnson AM, Keeney S, Siegel A, Sanford LD, Singletary SJ, Lonart G. Low (20 cGy) Doses of 1 GeV/u56Fe-Particle Radiation Lead to a Persistent Reduction in the Spatial Learning Ability of Rats. Radiat Res 2012; 177:146-51. [DOI: 10.1667/rr2637.1] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
33
|
Poulose SM, Bielinski DF, Carrihill-Knoll K, Rabin BM, Shukitt-Hale B. Exposure to 16O-particle radiation causes aging-like decrements in rats through increased oxidative stress, inflammation and loss of autophagy. Radiat Res 2011; 176:761-9. [PMID: 21962006 DOI: 10.1667/rr2605.1] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Exposing young rats to particles of high energy and charge (HZE particles), a ground-based model for exposure to cosmic rays, enhances indices of oxidative stress and inflammation, disrupts the functioning of neuronal communication, and alters cognitive behaviors. Even though exposure to HZE particles occurs at low fluence rates, the cumulative effects of long-term exposure result in molecular changes similar to those seen in aged animals. In the present study, we assessed markers of autophagy, a dynamic process for intracellular degradation and recycling of toxic proteins and organelles, as well as stress and inflammatory responses, in the brains of Sprague-Dawley rats irradiated at 2 months of age with 5 and 50 cGy and 1 Gy of ionizing oxygen particles ((16)O) (1000 MeV/n). Compared to nonirradiated controls, exposure to (16)O particles significantly inhibited autophagy function in the hippocampus as measured by accumulation of ubiquitin inclusion bodies such as P62/SQSTM1, autophagosome marker microtubule-associated protein 1 beta light chain 3 (MAP1B-LC3), beclin1 and proteins such as mammalian target of rapamycin (mTOR). The molecular changes measured at short (36 h) and long (75 days) intervals after (16)O-particle exposure indicate that the loss of autophagy function occurred shortly after exposure but was recovered via inhibition of mTOR. However, HZE-particle radiation caused significant sustained loss of protein kinase C alpha (PKC-α), a key G protein modulator involved in neuronal survival and functions of neuronal trophic factors. Exposure to (16)O particles also caused substantial increases in the levels of nuclear factor kappa B (NF-κB) and glial fibrillary acidic protein (GFAP), indicating glial cell activation 75 days after exposure. This is the first report to show the molecular effects of (16)O-particle radiation on oxidative stress, inflammation and loss of autophagy in the brain of young rats.
Collapse
Affiliation(s)
- Shibu M Poulose
- USDA-ARS, Human Nutrition Research Center on Aging at Tufts University, Boston, Massachusetts 02111, USA
| | | | | | | | | |
Collapse
|