1
|
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: 0.5] [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
|
2
|
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
|
3
|
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
|
4
|
Brand MD. Riding the tiger - physiological and pathological effects of superoxide and hydrogen peroxide generated in the mitochondrial matrix. Crit Rev Biochem Mol Biol 2020; 55:592-661. [PMID: 33148057 DOI: 10.1080/10409238.2020.1828258] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Elevated mitochondrial matrix superoxide and/or hydrogen peroxide concentrations drive a wide range of physiological responses and pathologies. Concentrations of superoxide and hydrogen peroxide in the mitochondrial matrix are set mainly by rates of production, the activities of superoxide dismutase-2 (SOD2) and peroxiredoxin-3 (PRDX3), and by diffusion of hydrogen peroxide to the cytosol. These considerations can be used to generate criteria for assessing whether changes in matrix superoxide or hydrogen peroxide are both necessary and sufficient to drive redox signaling and pathology: is a phenotype affected by suppressing superoxide and hydrogen peroxide production; by manipulating the levels of SOD2, PRDX3 or mitochondria-targeted catalase; and by adding mitochondria-targeted SOD/catalase mimetics or mitochondria-targeted antioxidants? Is the pathology associated with variants in SOD2 and PRDX3 genes? Filtering the large literature on mitochondrial redox signaling using these criteria highlights considerable evidence that mitochondrial superoxide and hydrogen peroxide drive physiological responses involved in cellular stress management, including apoptosis, autophagy, propagation of endoplasmic reticulum stress, cellular senescence, HIF1α signaling, and immune responses. They also affect cell proliferation, migration, differentiation, and the cell cycle. Filtering the huge literature on pathologies highlights strong experimental evidence that 30-40 pathologies may be driven by mitochondrial matrix superoxide or hydrogen peroxide. These can be grouped into overlapping and interacting categories: metabolic, cardiovascular, inflammatory, and neurological diseases; cancer; ischemia/reperfusion injury; aging and its diseases; external insults, and genetic diseases. Understanding the involvement of mitochondrial matrix superoxide and hydrogen peroxide concentrations in these diseases can facilitate the rational development of appropriate therapies.
Collapse
|
5
|
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: 3.4] [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
|
6
|
Schreurs AS, Torres S, Truong T, Moyer EL, Kumar A, Tahimic CGT, Alwood JS, Globus RK. Skeletal tissue regulation by catalase overexpression in mitochondria. Am J Physiol Cell Physiol 2020; 319:C734-C745. [PMID: 32783660 DOI: 10.1152/ajpcell.00068.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Accumulation of oxidative damage from excess reactive oxygen species (ROS) may contribute to skeletal aging and mediate adverse responses to physiological challenges. Wild-type (WT) mice and transgenic mice (male, 16 wk of age) with human catalase targeted to the mitochondria (mCAT) were analyzed for skeletal responses to the remodeling stimuli of combined hind-limb unloading and exposure to ionizing radiation (137Cs, 2 Gy). Treatment for 2 wk caused lipid peroxidation in the bones WT but not mCAT mice, showing that transgene expression mitigated oxidative stress. Ex vivo osteoblast colony growth rate was 95% greater in mCAT than WT mice and correlated with catalase activity levels (P < 0.005, r = 0.67), although terminal osteoblast and osteoclast differentiation were unaffected. mCAT mice had lower cancellous bone volume and cortical size than WT mice. Ambulatory control mCAT animals also displayed reduced cancellous and cortical structural properties compared with control WT mice. In mCAT but not WT mice, treatment caused an unexpectedly rapid radial expansion (+8% cortical area, +22% moment of inertia), reminiscent of compensatory bone growth during advancing age. In contrast, treatment caused similar structural deficits in cancellous tissue of mCAT and WT mice. In sum, mitochondrial ROS signaling via H2O2 was important for the acquisition of adult bone structure and catalase overexpression failed to protect cancellous tissue from treatment. In contrast, catabolic stimuli caused radial expansion in mCAT not WT mice, suggesting that mitochondrial ROS in skeletal cells act to suppress tissue turnover in response to remodeling challenges.
Collapse
Affiliation(s)
- Ann-Sofie Schreurs
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, California.,Universities Space Research Association, Moffett Field, California
| | - Samantha Torres
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, California.,Blue Marble Space Institute of Science, Seattle, Washington
| | - Tiffany Truong
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, California.,Blue Marble Space Institute of Science, Seattle, Washington
| | - Eric L Moyer
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, California.,Blue Marble Space Institute of Science, Seattle, Washington
| | - Akhhilesh Kumar
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, California.,Universities Space Research Association, Moffett Field, California
| | - Candice G T Tahimic
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, California.,KBR, Moffett Field, California
| | - Joshua S Alwood
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, California
| | - Ruth K Globus
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, California
| |
Collapse
|
7
|
Lv X, He H, Yang Y, Han L, Guo Z, Chen H, Li J, Qiu Y, Xie C. Radiation-induced hippocampal atrophy in patients with nasopharyngeal carcinoma early after radiotherapy: a longitudinal MR-based hippocampal subfield analysis. Brain Imaging Behav 2020; 13:1160-1171. [PMID: 30054872 DOI: 10.1007/s11682-018-9931-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Increasing evidence indicates that radiation-induced injury to the hippocampus may play a critical role in neurocognitive dysfunction in patients with nasopharyngeal carcinoma (NPC). However, few studies have assessed RT-induced hippocampal structural alterations in these patients early after radiotherapy (RT). In this study, 58 NPC patients were longitudinally followed up prior to treatment initiation as well as 3 and 6 months after RT, respectively. Twenty comparable normal controls were recruited and followed up in parallel. A novel magnetic resonance imaging (MRI)-based automated method was used to label hippocampal subfields. The linear mixed model was employed to evaluate longitudinal changes in the volumes of the whole hippocampus and seven hippocampal subfields. Time-dependent volume reduction was observed in the bilateral hippocampus, as well as in the bilateral granule cell layer (GCL), bilateral cornu ammonis 1 (CA1), bilateral molecular layer (ML), and bilateral subiculum (SUB) in NPC patients, but not in controls. Moreover, volume deficits in the bilateral hippocampus, bilateral GCL, and right ML showed dose-dependent patterns, and high volume losses in the bilateral hippocampus, bilateral GCL, left SUB, and right ML were associated with a rapid decline in cognitive function. Our findings demonstrated that the hippocampal subfields were selectively injured by irradiation-related early neurotoxic effects, which might account for cognitive impairment in NPC patients at an early stage after RT. Further, structural MRI could serve as a potential noninvasive imaging biomarker for the early detection of radiation effects on the hippocampus in NPC patients after RT.
Collapse
Affiliation(s)
- Xiaofei Lv
- Department of Medical Imaging, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, People's Republic of China
| | - Haoqiang He
- Department of Medical Imaging, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, People's Republic of China
| | - Yadi Yang
- Department of Medical Imaging, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, People's Republic of China
| | - Lujun Han
- Department of Medical Imaging, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, People's Republic of China
| | - Zheng Guo
- Department of Oncology, The First Affiliated Hospital of Ganzhou Medical University, Ganzhou, Jiangxi, People's Republic of China
| | - Hong Chen
- Department of Medical Imaging, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, People's Republic of China
| | - Jing Li
- Department of Medical Imaging, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, People's Republic of China
| | - Yingwei Qiu
- Department of Radiology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, People's Republic of China.
| | - Chuanmiao Xie
- Department of Medical Imaging, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, People's Republic of China.
| |
Collapse
|
8
|
Smith SM, Giedzinski E, Angulo MC, Lui T, Lu C, Park AL, Tang S, Martirosian V, Ru N, Chmielewski NN, Liang Y, Baulch JE, Acharya MM, Limoli CL. Functional equivalence of stem cell and stem cell-derived extracellular vesicle transplantation to repair the irradiated brain. Stem Cells Transl Med 2020; 9:93-105. [PMID: 31568685 PMCID: PMC6954724 DOI: 10.1002/sctm.18-0227] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 06/17/2019] [Indexed: 01/22/2023] Open
Abstract
Cranial radiotherapy, although beneficial for the treatment of brain tumors, inevitably leads to normal tissue damage that can induce unintended neurocognitive complications that are progressive and debilitating. Ionizing radiation exposure has also been shown to compromise the structural integrity of mature neurons throughout the brain, an effect believed to be at least in part responsible for the deterioration of cognitive health. Past work has shown that cranially transplanted human neural stem cells (hNSCs) or their extracellular vesicles (EVs) afforded long-term beneficial effects on many of these cognitive decrements. To provide additional insight into the potential neuroprotective mechanisms of cell-based regenerative strategies, we have analyzed hippocampal neurons for changes in structural integrity and synaptic remodeling after unilateral and bilateral transplantation of hNSCs or EVs derived from those same cells. Interestingly, hNSCs and EVs similarly afforded protection to host neurons, ameliorating the impact of irradiation on dendritic complexity and spine density for neurons present in both the ipsilateral and contralateral hippocampi 1 month following irradiation and transplantation. These morphometric improvements were accompanied by increased levels of glial cell-derived growth factor and significant attenuation of radiation-induced increases in postsynaptic density protein 95 and activated microglia were found ipsi- and contra-lateral to the transplantation sites of the irradiated hippocampus treated with hNSCs or hNSC-derived EVs. These findings document potent far-reaching neuroprotective effects mediated by grafted stem cells or EVs adjacent and distal to the site of transplantation and support their potential as therapeutic agents to counteract the adverse effects of cranial irradiation.
Collapse
Affiliation(s)
- Sarah M. Smith
- Department of Radiation OncologyUniversity of CaliforniaIrvineCalifornia
| | - Erich Giedzinski
- Department of Radiation OncologyUniversity of CaliforniaIrvineCalifornia
| | - Maria C. Angulo
- Department of Radiation OncologyUniversity of CaliforniaIrvineCalifornia
| | - Tiffany Lui
- Department of Radiation OncologyUniversity of CaliforniaIrvineCalifornia
| | - Celine Lu
- Department of Radiation OncologyUniversity of CaliforniaIrvineCalifornia
| | - Audrey L. Park
- Department of Radiation OncologyUniversity of CaliforniaIrvineCalifornia
| | - Sharon Tang
- Department of Radiation OncologyUniversity of CaliforniaIrvineCalifornia
| | - Vahan Martirosian
- Department of Radiation OncologyUniversity of CaliforniaIrvineCalifornia
| | - Ning Ru
- Department of Radiation OncologyUniversity of CaliforniaIrvineCalifornia
| | | | - Yaxuan Liang
- Department of Radiation OncologyUniversity of CaliforniaIrvineCalifornia
| | - Janet E. Baulch
- Department of Radiation OncologyUniversity of CaliforniaIrvineCalifornia
| | - Munjal M. Acharya
- Department of Radiation OncologyUniversity of CaliforniaIrvineCalifornia
| | - Charles L. Limoli
- Department of Radiation OncologyUniversity of CaliforniaIrvineCalifornia
| |
Collapse
|
9
|
Kiffer F, Boerma M, Allen A. Behavioral effects of space radiation: A comprehensive review of animal studies. LIFE SCIENCES IN SPACE RESEARCH 2019; 21:1-21. [PMID: 31101151 PMCID: PMC7150604 DOI: 10.1016/j.lssr.2019.02.004] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 02/14/2019] [Accepted: 02/17/2019] [Indexed: 05/04/2023]
Abstract
As NASA prepares for the first manned mission to Mars in the next 20 years, close attention has been placed on the cognitive welfare of astronauts, who will likely endure extended durations in confinement and microgravity and be subjected to the radioactive charged particles travelling at relativistic speeds in interplanetary space. The future of long-duration manned spaceflight, thus, depends on understanding the individual hazards associated with the environment beyond Earth's protective magnetosphere. Ground-based single-particle studies of exposed mice and rats have, in the last 30 years, overwhelmingly reported deficits in their cognitive behaviors. However, as particle-accelerator technologies at NASA's Space Radiation Laboratory continue to progress, more realistic representations of space radiation are materializing, including multiple-particle exposures and, eventually, at multiple energy distributions. These advancements help determine how to best mitigate possible hazards due to space radiation. However, risk models will depend on delineating which particles are most responsible for specific behavioral outcomes and whether multiple-particle exposures produce synergistic effects. Here, we review the literature on animal exposures by particle, energy, and behavioral assay to inform future mixed-field radiation studies of possible behavioral outcomes.
Collapse
Affiliation(s)
- Frederico Kiffer
- Division of Radiation Health, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, United States; Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, United States.
| | - Marjan Boerma
- Division of Radiation Health, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, United States; Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, United States.
| | - Antiño Allen
- Division of Radiation Health, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, United States; Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, United States; Department of Neurobiology & Developmental Sciences, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States.
| |
Collapse
|
10
|
Ahn JH, Shin BN, Song M, Kim H, Park JH, Lee TK, Park CW, Park YE, Lee JC, Yong JH, Lee CH, Hwang IK, Won MH, Lee YL. Intermittent fasting increases the expressions of SODs and catalase in granule and polymorphic cells and enhances neuroblast dendrite complexity and maturation in the adult gerbil dentate gyrus. Mol Med Rep 2019; 19:1721-1727. [PMID: 30628688 PMCID: PMC6390044 DOI: 10.3892/mmr.2019.9822] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 12/31/2018] [Indexed: 11/06/2022] Open
Abstract
Intermittent fasting (ImF) is known to reduce oxidative stress and affects adult neurogenesis in the hippocampal dentate gyrus. However, it is unknown how ImF affects endogenous antioxidants expressions, cell proliferation, and neuroblast differentiation and their dendrite remodeling over 3 months in the dentate gyrus of adult gerbils. The present study subjected 6‑month old male gerbils to a normal diet or alternate‑day ImF for 1, 2 and 3 months. Changes in body weight were not significantly different between gerbils fed a normal diet and on ImF. The present study also investigated the effects of ImF on antioxidant enzymes [superoxide dismutase (SOD)‑1, SOD2 and catalase] using immunohistochemistry, and endogenous cell proliferation, neuroblast differentiation and neuroblast dendrite complexity by using Ki67 (a cell proliferation marker) and doublecortin (neuroblast differentiation marker) immunohistochemistry in the dentate gyrus. SOD1, SOD2 and CAT immunoreactivities were shown in cells in the granule cell and polymorphic layers. SOD1, SOD2 and catalase immunoreactivity in the cells peaked at 2, 1 and 1 month, respectively, following ImF. Cell proliferation was ~250, 129 and 186% of the control, at 1, 2 and 3 months of ImF, respectively. Neuroblast differentiation was ~41, 32 and 12% of the control, at 1, 2 and 3 months of ImF, respectively, indicating that dendrites of neuroblasts were more arborized and developed at 3 months of ImF. Taken together, these results indicate that ImF for 3 months improves endogenous SOD1, SOD2 and catalase expressions and enhances cell proliferation, and neuroblast dendrites complexity and maturation in the adult gerbil dentate gyrus.
Collapse
Affiliation(s)
- Ji Hyeon Ahn
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| | - Bich Na Shin
- Danchunok Company, Chuncheon, Gangwon 24210, Republic of Korea
| | - Minah Song
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Hyunjung Kim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Joon Ha Park
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| | - Tae-Kyeong Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Cheol Woo Park
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Young Eun Park
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Jae-Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Jun-Hwan Yong
- Department of Occupational Therapy, Dongnam Health University, Suwon, Gyeonggi 16238, Republic of Korea
| | - Choong-Hyun Lee
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan, Chungcheongnam 31116, Republic of Korea
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Yun Lyul Lee
- Department of Physiology, College of Medicine and Institute of Neurodegeneration and Neuroregeneration, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| |
Collapse
|
11
|
Oxidative Stress as Cause, Consequence, or Biomarker of Altered Female Reproduction and Development in the Space Environment. Int J Mol Sci 2018; 19:ijms19123729. [PMID: 30477143 PMCID: PMC6320872 DOI: 10.3390/ijms19123729] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 11/12/2018] [Accepted: 11/20/2018] [Indexed: 12/22/2022] Open
Abstract
Oxidative stress has been implicated in the pathophysiology of numerous terrestrial disease processes and associated with morbidity following spaceflight. Furthermore, oxidative stress has long been considered a causative agent in adverse reproductive outcomes. The purpose of this review is to summarize the pathogenesis of oxidative stress caused by cosmic radiation and microgravity, review the relationship between oxidative stress and reproductive outcomes in females, and explore what role spaceflight-induced oxidative damage may have on female reproductive and developmental outcomes.
Collapse
|
12
|
Leavitt RJ, Limoli CL, Baulch JE. miRNA-based therapeutic potential of stem cell-derived extracellular vesicles: a safe cell-free treatment to ameliorate radiation-induced brain injury. Int J Radiat Biol 2018; 95:427-435. [PMID: 30252569 DOI: 10.1080/09553002.2018.1522012] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE This review compiles what is known about extracellular vesicles (EVs), their bioactive cargo, and how they might be used to treat radiation-induced brain injury. Radiotherapy (RT) is effective in cancer treatment, but can cause substantial damage to normal central nervous system tissue. Stem cell therapy has been shown to be effective in treating cognitive dysfunction arising from RT, but there remain safety concerns when grafting foreign stem cells into the brain (i.e. immunogenicity, teratoma). These limitations prompted the search for cell-free alternatives, and pointed to EVs that have been shown to have similar ameliorating effects in other tissues and injury models. CONCLUSIONS EVs are nano-scale and lipid-bound vesicles that readily pass the blood-brain barrier. Arguably the most important bioactive cargo within EVs are RNAs, in particular microRNAs (miRNA). A single miRNA can modulate entire gene networks and signalling within the recipient cell. Determining functionally relevant miRNA could lead to therapeutic treatments where synthetically-derived EVs are used as delivery vectors for miRNA. Stem cell-derived EVs can be effective in treating brain injury including radiation-induced cognitive deficits. Of particular interest are systemic modes of administration which obviate the need for invasive procedures.
Collapse
Affiliation(s)
- Ron J Leavitt
- a Department of Radiation Oncology , University of California Irvine , Irvine , CA , USA
| | - Charles L Limoli
- a Department of Radiation Oncology , University of California Irvine , Irvine , CA , USA
| | - Janet E Baulch
- a Department of Radiation Oncology , University of California Irvine , Irvine , CA , USA
| |
Collapse
|
13
|
Grosshans DR, Duman JG, Gaber MW, Sawakuchi G. Particle Radiation Induced Neurotoxicity in the Central Nervous System. Int J Part Ther 2018; 5:74-83. [PMID: 31773021 DOI: 10.14338/ijpt-18-00026.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 08/16/2018] [Indexed: 11/21/2022] Open
Abstract
For patients with primary or metastatic brain tumors, radiation therapy plays a central role in treatment. However, despite its efficacy, cranial radiation is associated with a range of side effects ranging from mild cognitive impairment to overt brain necrosis. Given the negative effects on patient quality of life, radiation-induced neurotoxicities have been the subject of intense study for decades. Photon-based therapy has been and largely remains the standard of care for the treatment of brain tumors. This is particularly true for patients with metastatic tumors who may need treatment to the whole brain or those with very aggressive tumors and a limited life expectancy. Particle therapy is now becoming more widely available for clinical use with the two most common particles used being protons and carbon ions. For patients with favorable prognoses, particularly childhood brain tumors, proton therapy is increasingly used for treatment. This is, in part, driven by the desire to reduce the potential for radiation-induced side effects, including lasting cognitive impairment, which may potentially be achieved by reducing dose to normal tissues using the unique physical properties of particle therapy. There is also interest in using carbon ion therapy for the treatment of aggressive brain tumors, as this form of particle therapy not only spares normal tissues but may also improve tumor control. The biological effects of particle therapy, both proton and carbon, may differ substantially from those of photon radiation. In this review, we briefly describe the unique physical properties of particle therapy that produce differential biological effects. Focusing on the effects of various radiation types on brain parenchyma, we then describe biological effects and potential mechanisms underlying these, comparing to photon studies and highlighting potential clinical implications.
Collapse
Affiliation(s)
- David R Grosshans
- Departments of Radiation and Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joseph G Duman
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - M Waleed Gaber
- Department of Pediatrics, Texas Children's Cancer Center, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Gabriel Sawakuchi
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
14
|
Cacao E, Parihar VK, Limoli CL, Cucinotta FA. Stochastic Modeling of Radiation-induced Dendritic Damage on in silico Mouse Hippocampal Neurons. Sci Rep 2018; 8:5494. [PMID: 29615729 PMCID: PMC5882641 DOI: 10.1038/s41598-018-23855-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 03/21/2018] [Indexed: 12/20/2022] Open
Abstract
Cognitive dysfunction associated with radiotherapy for cancer treatment has been correlated to several factors, one of which is changes to the dendritic morphology of neuronal cells. Alterations in dendritic geometry and branching patterns are often accompanied by deficits that impact learning and memory. The purpose of this study is to develop a novel predictive model of neuronal dendritic damages caused by exposure to low linear energy transfer (LET) radiation, such as X-rays, γ-rays and high-energy protons. We established in silico representations of mouse hippocampal dentate granule cell layer (GCL) and CA1 pyramidal neurons, which are frequently examined in radiation-induced cognitive decrements. The in silico representations are used in a stochastic model that describes time dependent dendritic damage induced by exposure to low LET radiation. Changes in morphometric parameters, such as total dendritic length, number of branch points and branch number, including the Sholl analysis for single neurons are described by the model. Our model based predictions for different patterns of morphological changes based on energy deposition in dendritic segments (EDDS) will serve as a useful basis to compare specific patterns of morphological alterations caused by EDDS mechanisms.
Collapse
Affiliation(s)
- Eliedonna Cacao
- Department of Health Physics and Diagnostic Sciences, University of Nevada, Las Vegas, NV, United States of America
| | - Vipan K Parihar
- Department of Radiation Oncology, University of California, Irvine, CA, United States of America
| | - Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine, CA, United States of America
| | - Francis A Cucinotta
- Department of Health Physics and Diagnostic Sciences, University of Nevada, Las Vegas, NV, United States of America.
| |
Collapse
|
15
|
Alp M, Cucinotta FA. Biophysics Model of Heavy-Ion Degradation of Neuron Morphology in Mouse Hippocampal Granular Cell Layer Neurons. Radiat Res 2018; 189:312-325. [PMID: 29502499 PMCID: PMC5872156 DOI: 10.1667/rr14923.1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Exposure to heavy-ion radiation during cancer treatment or space travel may cause cognitive detriments that have been associated with changes in neuron morphology and plasticity. Observations in mice of reduced neuronal dendritic complexity have revealed a dependence on radiation quality and absorbed dose, suggesting that microscopic energy deposition plays an important role. In this work we used morphological data for mouse dentate granular cell layer (GCL) neurons and a stochastic model of particle track structure and microscopic energy deposition (ED) to develop a predictive model of high-charge and energy (HZE) particle-induced morphological changes to the complex structures of dendritic arbors. We represented dendrites as cylindrical segments of varying diameter with unit aspect ratios, and developed a fast sampling method to consider the stochastic distribution of ED by δ rays (secondary electrons) around the path of heavy ions, to reduce computational times. We introduce probabilistic models with a small number of parameters to describe the induction of precursor lesions that precede dendritic snipping, denoted as snip sites. Predictions for oxygen (16O, 600 MeV/n) and titanium (48Ti, 600 MeV/n) particles with LET of 16.3 and 129 keV/μm, respectively, are considered. Morphometric parameters to quantify changes in neuron morphology are described, including reduction in total dendritic length, number of branch points and branch numbers. Sholl analysis is applied for single neurons to elucidate dose-dependent reductions in dendritic complexity. We predict important differences in measurements from imaging of tissues from brain slices with single neuron cell observations due to the role of neuron death through both soma apoptosis and excessive dendritic length reduction. To further elucidate the role of track structure, random segment excision (snips) models are introduced and a sensitivity study of the effects of the modes of neuron death in predictions of morphometric parameters is described. An important conclusion of this study is that δ rays play a major role in neuron morphological changes due to the large spatial distribution of damage sites, which results in a reduced dependence on LET, including modest difference between 16O and 48Ti, compared to damages resulting from ED in localized damage sites.
Collapse
Affiliation(s)
- Murat Alp
- Department of Health Physics and Diagnostic Sciences, University of Nevada, Las Vegas, Las Vegas, Nevada
| | - Francis A. Cucinotta
- Department of Health Physics and Diagnostic Sciences, University of Nevada, Las Vegas, Las Vegas, Nevada
| |
Collapse
|
16
|
Proceedings of the National Cancer Institute Workshop on Charged Particle Radiobiology. Int J Radiat Oncol Biol Phys 2017; 100:816-831. [PMID: 29485053 DOI: 10.1016/j.ijrobp.2017.12.260] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 12/05/2017] [Accepted: 12/11/2017] [Indexed: 12/21/2022]
Abstract
In April 2016, the National Cancer Institute hosted a multidisciplinary workshop to discuss the current knowledge of the radiobiological aspects of charged particles used in cancer therapy to identify gaps in that knowledge that might hinder the effective clinical use of charged particles and to propose research that could help fill those gaps. The workshop was organized into 10 topics ranging from biophysical models to clinical trials and included treatment optimization, relative biological effectiveness of tumors and normal tissues, hypofractionation with particles, combination with immunotherapy, "omics," hypoxia, and particle-induced second malignancies. Given that the most commonly used charged particle in the clinic currently is protons, much of the discussion revolved around evaluating the state of knowledge and current practice of using a relative biological effectiveness of 1.1 for protons. Discussion also included the potential advantages of heavier ions, notably carbon ions, because of their increased biological effectiveness, especially for tumors frequently considered to be radiation resistant, increased effectiveness in hypoxic cells, and potential for differentially altering immune responses. The participants identified a large number of research areas in which information is needed to inform the most effective use of charged particles in the future in clinical radiation therapy. This unique form of radiation therapy holds great promise for improving cancer treatment.
Collapse
|
17
|
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: 2.5] [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
|
18
|
Tahimic CGT, Globus RK. Redox Signaling and Its Impact on Skeletal and Vascular Responses to Spaceflight. Int J Mol Sci 2017; 18:ijms18102153. [PMID: 29035346 PMCID: PMC5666834 DOI: 10.3390/ijms18102153] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Revised: 09/30/2017] [Accepted: 10/10/2017] [Indexed: 12/16/2022] Open
Abstract
Spaceflight entails exposure to numerous environmental challenges with the potential to contribute to both musculoskeletal and vascular dysfunction. The purpose of this review is to describe current understanding of microgravity and radiation impacts on the mammalian skeleton and associated vasculature at the level of the whole organism. Recent experiments from spaceflight and ground-based models have provided fresh insights into how these environmental stresses influence mechanisms that are related to redox signaling, oxidative stress, and tissue dysfunction. Emerging mechanistic knowledge on cellular defenses to radiation and other environmental stressors, including microgravity, are useful for both screening and developing interventions against spaceflight-induced deficits in bone and vascular function.
Collapse
Affiliation(s)
- Candice G T Tahimic
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA.
- KBRWyle, Moffett Field, CA 94035, USA.
| | - Ruth K Globus
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA.
| |
Collapse
|
19
|
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: 4.7] [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
|
20
|
Limoli CL. Understanding and targeting dynamic stress responses of the brain: What we have learned and how to improve neurocognitive outcome following neurotoxic insult. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2016; 57:319-321. [PMID: 27208487 DOI: 10.1002/em.22022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 04/28/2016] [Indexed: 06/05/2023]
Affiliation(s)
- Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine, California
| |
Collapse
|