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
|
Wuyts FL, Deblieck C, Vandevoorde C, Durante M. Brains in space: impact of microgravity and cosmic radiation on the CNS during space exploration. Nat Rev Neurosci 2025; 26:354-371. [PMID: 40247135 DOI: 10.1038/s41583-025-00923-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2025] [Indexed: 04/19/2025]
Abstract
Solar system exploration is a grand endeavour of humankind. Space agencies have been planning crewed missions to the Moon and Mars for several decades. However, several environmental stress factors in space, such as microgravity and cosmic radiation, confer health risks for human explorers. This Review examines the effects of microgravity and exposure to cosmic radiation on the CNS. Microgravity presents challenges for the brain, necessitating the development of adaptive movement and orientation strategies to cope with alterations in sensory information. Exposure to microgravity also affects cognitive function to a certain extent. Recent MRI results show that microgravity affects brain structure and function. Post-flight recovery from these changes is gradual, with some lasting up to a year. Regarding cosmic radiation, animal experiments suggest that the brain could be much more sensitive to this stressor than may be expected from experiences on Earth. This may be due to the presence of energetic heavy ions in space that have an impact on cognitive function, even at low doses. However, all data about space radiation risk stem from rodent experiments, and extrapolation of these data to humans carries a high degree of uncertainty. Here, after presenting an overview of current knowledge in the above areas, we provide a concise description of possible counter-measures to protect the brain against microgravity and cosmic radiation during future space missions.
Collapse
Affiliation(s)
- Floris L Wuyts
- Laboratory for Equilibrium Investigations and Aerospace (LEIA), University of Antwerp, Antwerp, Belgium
| | - Choi Deblieck
- Laboratory for Equilibrium Investigations and Aerospace (LEIA), University of Antwerp, Antwerp, Belgium
| | - Charlot Vandevoorde
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Marco Durante
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany.
- Institute for Condensed Matter of Physics, Technische Universität Darmstadt, Darmstadt, Germany.
- Department of Physics 'Ettore Pancini', University Federico II, Naples, Italy.
| |
Collapse
|
3
|
Chang HHV, Kim G, Kim KS, Britten RA. Reduced set-shifting processing speed in male rats following low dose (10 cGy) proton exposure. LIFE SCIENCES IN SPACE RESEARCH 2025; 45:137-142. [PMID: 40280634 DOI: 10.1016/j.lssr.2025.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 02/12/2025] [Accepted: 02/17/2025] [Indexed: 04/29/2025]
Abstract
Space radiation (SR) exposure poses significant biomedical risks, including effects on the central nervous system (CNS). These risks are particularly relevant to cognitive function during long-duration space missions. One critical cognitive skill is decision-making, which requires attentional set-shifting (ATSET)-the ability to quickly assess problems, evaluate options, and select the best actions. Previous studies have shown that exposure to <10 cGy of SR ions impairs ATSET performance in animal models. However, the impact of low LET (< 1 keV/μm) protons, which significantly contribute to the total radiation flux astronauts encounter within spacecraft, on ATSET performance is unknown. To address this gap, we evaluated the effects of cranial irradiation with 10 cGy of 100 MeV/n protons (LET = 0.732 keV/μm) on ATSET performance in male Sprague-Dawley rats. We also investigated whether concurrent exposure to variable gravity (hypergravity step-up, step down, purported to have the same effect as exposure to microgravity (another major spaceflight stressor) exacerbated SR-induced cognitive deficits. Our findings indicate that proton exposure alone significantly impaired ATSET performance, as evidenced by decreased processing speed while performing compound discrimination reversal and extra-dimensional shifting. Notably, no additive or synergistic effects were observed when hypergravity was combined with proton exposure. The impact that low-dose proton exposure has on CNS functionality, particularly in reducing processing speed during complex tasks, warrant further investigation. If similar cognitive deficits were to occur in astronauts exposed to galactic cosmic rays, mission success and safety could be significantly compromised.
Collapse
Affiliation(s)
| | - Gyutae Kim
- Research Institute for Aerospace Medicine, Inha University, Incheon, Korea
| | - Kyu-Sung Kim
- Research Institute for Aerospace Medicine, Inha University, Incheon, Korea; Department of Otolaryngology Head and Neck Surgery, Inha University Hospital, Incheon, Korea
| | - Richard A Britten
- Radiation Oncology, USA; Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Macon and Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA
| |
Collapse
|
4
|
Straume T, Mora AM, Brown JB, Bansal I, Rabin BM, Braby LA, Wyrobek AJ. Non-DNA radiosensitive targets that initiate persistent behavioral deficits in rats exposed to space radiation. LIFE SCIENCES IN SPACE RESEARCH 2025; 45:44-60. [PMID: 40280642 DOI: 10.1016/j.lssr.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 12/16/2024] [Accepted: 12/20/2024] [Indexed: 04/29/2025]
Abstract
Predicting future CNS risks for astronauts during deep-space missions will rely substantially on ground-based rodent data with space-relevant ions and behaviors. For rats, the accumulated evidence indicates that less densely ionizing radiation, such as 4He and 12C ions, induce behavior deficits at lower doses than densely ionizing ions, such as 48Ti and 56Fe. However, this observation conflicts with standard somatic radiobiology, in which densely ionizing ions are generally more effective than less densely ionizing ions, and where the DNA/nucleus is the accepted target for radiation-induced tumorigenesis, cytogenetic aberrations, genetic mutations, and reproductive cell death. To gain deeper insight into the subcellular nature of the radiation targets for behavior risks, we compared the effects of dose, fluence, and linear energy transfer (LET) of 4He and 56Fe particles using existing datasets for four distinct behavioral outcomes in rats: elevated plus maze (EPM-anxiety), novel object recognition (NOR-memory), operant responding (OR-response to environmental stimuli), and attentional set-shifting (ATSET-cognitive flexibility). We confirmed that less densely ionizing particles (except protons) showed ∼100-fold lower threshold doses than densely ionizing particles for behavioral deficits (0.1-1 cGy for 4He vs. 15-100 cGy for 56Fe). However, when analyzed by fluence the behavioral responses converged, indicating that 4He and 56Fe were equally effective on a per-track basis. When analyzed by LET, there were ∼100-fold differences in the LET for maximum effectiveness for behavioral deficits and DNA endpoints (∼1 vs ∼100 keV/μm, respectively). These unique features of radiation-induced behavioral deficits (high sensitivity to particles in the 1-keV/μm range, insensitivity to protons in the 0.2 keV/μm range, and isofluence dependence for particles with LET>1 keV/μm) provide evidence in support of a new hypothesis of sub-micron sized radiosensitive targets for behavioral effects consistent with the thickness of plasma membranes and/or small subcellular structures, smaller than a whole synapse. Like our behavior findings, mouse immature oocyte killing which is known to have a plasma membrane target was also better explained by fluence, rather than dose. In contrast, fluence analyses for DNA/nuclear endpoints in somatic cells (e.g., tumor induction, chromosome aberrations) showed opposite results, suggesting that behavior targets are not DNA. Our findings raise questions regarding the identity of subcellular targets and the multi-cellular functional unit for behavior risks, low-dose susceptibility, and generalizability from rat to other species and astronauts.
Collapse
Affiliation(s)
- Tore Straume
- Lawrence Berkeley National Laboratory, University of California, 1 Cyclotron Road, Berkeley, CA 94720, USA; NASA Ames Research Center (retired affiliation), Moffett Field, CA 94035, USA.
| | - Ana M Mora
- School of Public Health, University of California, Berkeley, CA 94720, USA
| | - James B Brown
- Lawrence Berkeley National Laboratory, University of California, 1 Cyclotron Road, Berkeley, CA 94720, USA
| | - Ishan Bansal
- Lawrence Berkeley National Laboratory, University of California, 1 Cyclotron Road, Berkeley, CA 94720, USA
| | | | | | - Andrew J Wyrobek
- Lawrence Berkeley National Laboratory, University of California, 1 Cyclotron Road, Berkeley, CA 94720, USA.
| |
Collapse
|
5
|
Yan L, Er T, Sun S, Deng Y, Wan Z, Zhao J, Wang A, Liu B, Wang Q, Sui L, Ma H. Distinct astrocyte activation patterns associated with neuroinflammation induced by gamma and proton beam irradiation. Sci Rep 2025; 15:11481. [PMID: 40181058 PMCID: PMC11968924 DOI: 10.1038/s41598-025-94812-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 03/17/2025] [Indexed: 04/05/2025] Open
Abstract
The aim of this study was to investigate the impact of radiation exposure on astrocyte response and assess their potential roles and mechanisms in surrounding neural cells. Healthy male rats were irradiated different radiation types to induce the neural inflammation. U87-MG cells were exposed respectively to gamma rays (2 Gy and 10 Gy) and proton irradiation (0.1 Gy and 0.5 Gy). Cell viability, mRNA expression, mitochondrial membrane potential, glucose uptake and cytokine levels were analyzed respectively to evaluate the neuroinflammation or neural damage. Gamma rays and proton beam irradiation induced distinct patterns of inflammatory factor expression in the hippocampal region of rats. Moreover, we observed changes in cell morphology and a dose-dependent inhibition of cell proliferation across all radiation types. Significant upregulation of caspase-8 and caspase-3 enzymatic activities in U87-MG cells was observed after exposure to gamma rays. Astrocytes showed increased expression of GFAP, C3, and PTX3 after exposure to gamma rays, and downregulation while exposure to proton. Additionally, proton beam irradiation potentially increased glutamine synthesis in astrocytes. Furthermore, we investigated the influence of irradiated astrocytes on neurons via mitochondrial integrity, neurotransmitter levels, and glucose metabolism. Additionally, the expression of miR92a-3p, which can significantly downregulate GFAP and IL-6 expression, was downregulated by gamma rays, while upregulated by proton irradiation. The findings highlight the differential impact of gamma rays and proton radiation on inflammatory responses in vivo, with gamma rays inducing a pro-inflammatory effect and proton radiation exerting anti-inflammatory properties. Overall, this study provides valuable insights for radiotherapy management.
Collapse
Affiliation(s)
- Liben Yan
- School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China
| | - Tianyi Er
- School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China
| | - Shaoqian Sun
- School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China.
| | - Yulin Deng
- School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China
| | - Zhirong Wan
- Department of Neurology, Aerospace Center Hospital, Beijing, China
| | - Jing Zhao
- Department of Neurology, Aerospace Center Hospital, Beijing, China
| | - Ailu Wang
- School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China
| | - Beiqin Liu
- School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China
| | - Qiaojuan Wang
- Department of Nuclear Physics, China Institute of Atomic Energy, Beijing, 100081, China
| | - Li Sui
- Department of Nuclear Physics, China Institute of Atomic Energy, Beijing, 100081, China.
| | - Hong Ma
- School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China.
| |
Collapse
|
6
|
Smits E, Reid FE, Tamgue EN, Alvarado Arriaga P, Nguyen C, Britten RA. Sex-Dependent Changes in Risk-Taking Predisposition of Rats Following Space Radiation Exposure. Life (Basel) 2025; 15:449. [PMID: 40141792 PMCID: PMC11943666 DOI: 10.3390/life15030449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/21/2025] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
The Artemis missions will establish a sustainable human presence on the Moon, serving as a crucial steppingstone for future Mars exploration. Astronauts on these ambitious missions will have to successfully complete complex tasks, which will frequently involve rapid and effective decision making under unfamiliar or high-pressure conditions. Exposure to low doses of space radiation (SR) can impair key executive functions critical to decision making. This study examined the effects of exposure to 10 cGy of Galactic Cosmic Ray simulated radiation (GCRsim) on decision-making performance in male and female rats with a naturally low predisposition for risk-taking (RTP) prior to exposure. Rats were assessed at monthly intervals following SR exposure and the RTP performance contrasted with that observed during the prescreening process. Exposure to 10 cGy of GCRsim impaired decision making in both male and female rats, with sex-dependent outcomes. By 30 days after SR exposure, female rats became more risk-prone, making less profitable decisions, while male rats retained their decision-making strategies but took significantly longer to make selections. However, continued practice in the RTP tasks appeared to reduce/reverse these performance deficits. This study has expanded our understanding of the range of cognitive processes impacted by SR to include decision making.
Collapse
Affiliation(s)
- Elliot Smits
- EVMS School of Medicine, Macon and Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA; (E.S.); (F.E.R.)
| | - Faith E. Reid
- EVMS School of Medicine, Macon and Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA; (E.S.); (F.E.R.)
| | - Ella N. Tamgue
- EVMS Radiation Oncology, Eastern Virginia Medical School, Macon and Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA (P.A.A.); (C.N.)
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Macon and Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA
| | - Paola Alvarado Arriaga
- EVMS Radiation Oncology, Eastern Virginia Medical School, Macon and Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA (P.A.A.); (C.N.)
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Macon and Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA
| | - Charles Nguyen
- EVMS Radiation Oncology, Eastern Virginia Medical School, Macon and Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA (P.A.A.); (C.N.)
| | - Richard A. Britten
- EVMS Radiation Oncology, Eastern Virginia Medical School, Macon and Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA (P.A.A.); (C.N.)
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Macon and Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA
| |
Collapse
|
7
|
Dickstein DL, Zhang R, Ru N, Vozenin MC, Perry BC, Wang J, Baulch JE, Acharya MM, Limoli CL. Structural plasticity of pyramidal cell neurons measured after FLASH and conventional dose-rate irradiation. Brain Struct Funct 2025; 230:41. [PMID: 40024988 PMCID: PMC11872753 DOI: 10.1007/s00429-025-02902-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 02/17/2025] [Indexed: 03/04/2025]
Abstract
Evidence shows that ultra-high dose-rate FLASH-radiotherapy (FLASH-RT) provides relative protection against normal tissue complications and functional decrements in the irradiated brain. Past work has shown that radiation-induced cognitive impairment, neuroinflammation and reduced structural complexity ofgranule cell neurons were not observed to the same extent after FLASH-RT (> MGy/s) compared to conventional dose-rate (CONV, 0.1 Gy/s) delivery. In this study, we explored the sensitivity of hippocampal CA1 and medial prefrontal cortex (mPFC) pyramidal neurons to cranial irradiation and dose-rate modulation using electron and confocal microscopy. Neuron ultrastructural analyses by electron microscopy after 10 Gy FLASH- or CONV-RT exposures indicated that irradiation had little impact on dendritic complexity and synapse density in the CA1, but did increase the length and head diameter of smaller non-perforated synapses. Similarly, irradiation caused no change in mPFC prelimbic/infralimbic axospinous synapse density, but reductions in non-perforated synapse diameters. While irradiation resulted in thinner myelin sheaths compared to controls, none of these metrics were dose-rate sensitive. Analysis of fluorescently labeled CA1 neurons revealed no radiation-induced or dose-rate-dependent changes in overall dendritic complexity or spine density, in contrast to our past analysis of granule cell neurons. Super-resolution confocal microscopy following a clinical dosing paradigm (3 × 10 Gy) showed significant reductions in excitatory vesicular glutamate transporter 1 and inhibitory vesicular GABA transporter puncta density within the CA1 that were largely dose-rate independent. Collectively, these data reveal that, compared to granule cell neurons, CA1 and mPFC neurons are relatively more radioresistant irrespective of radiation dose-rate.
Collapse
Affiliation(s)
- Dara L Dickstein
- Department of Pathology, Uniformed Services University of Health Sciences, Bethesda, MD, 20814, USA.
- The Henry M. Jackson Foundation for the Advancement of Military Health Inc, Bethesda, MD, USA.
| | - Richard Zhang
- Department of Radiation Oncology, University of California, Irvine School of Medicine, Irvine, CA, USA
| | - Ning Ru
- Department of Radiation Oncology, University of California, Irvine School of Medicine, Irvine, CA, USA
| | - Marie-Catherine Vozenin
- Secteur Radio-Oncologie et Radiobiologie, Hôpitaux Universitaires de Genève, Geneva, Switzerland
| | - Bayley C Perry
- Department of Pathology, Uniformed Services University of Health Sciences, Bethesda, MD, 20814, USA
- The Henry M. Jackson Foundation for the Advancement of Military Health Inc, Bethesda, MD, USA
| | - Juan Wang
- Department of Pathology, Uniformed Services University of Health Sciences, Bethesda, MD, 20814, USA
- The Henry M. Jackson Foundation for the Advancement of Military Health Inc, Bethesda, MD, USA
| | - Janet E Baulch
- Department of Radiation Oncology, University of California, Irvine School of Medicine, Irvine, CA, USA
| | - Munjal M Acharya
- Department of Radiation Oncology, University of California, Irvine School of Medicine, Irvine, CA, USA
- Department of Anatomy and Neurobiology, University of California, Irvine School of Medicine, Irvine, CA, USA
| | - Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine School of Medicine, Irvine, CA, USA.
| |
Collapse
|
8
|
Kokhan VS, Chaprov K, Abaimov DA, Nesterov MS, Pikalov VA. Combined irradiation by gamma-rays and carbon-12 nuclei caused hyperlocomotion and change in striatal metabolism of rats. LIFE SCIENCES IN SPACE RESEARCH 2025; 44:99-107. [PMID: 39864919 DOI: 10.1016/j.lssr.2024.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 07/11/2024] [Accepted: 08/15/2024] [Indexed: 01/28/2025]
Abstract
Exposure to ionizing radiation during manned deep space missions to Mars could lead to functional impairments of the central nervous system, which may compromise the success of the mission and affect the quality of life for returning astronauts. Along with radiation-induced changes in cognitive abilities and emotional status, the effects of increased motor activity were observed. The mechanisms behind these phenomena still remain unresolved. We conducted a study on grip strength, locomotor activity and intrasession habituation to novelty in 5-month-old rats after exposure to radiation (combined 0.4 Gy gamma-rays and 0.14 Gy 12C nuclei). At the same time, we carried out neurochemical and molecular analysis of the nucleus accumbens (NAc) and the dorsal striatum (dST). The study revealed radiation-induced hyperlocomotion and enhanced habituation. It also showed an increase in choline concentration and a decreased in 5-hydroxyindoleacetic acid concentration in the NAc after irradiation. In addition to this, a down-regulation of syntaxin 1A in NAc and dST as well as up-regulation α-synuclein in NAc were observed. The obtained data indicate both the damaging effect of irradiation on striatum tissues and the initiation of neuronal/axonal regeneration processes. It is hypothesized that the increase in choline concentration in NAc and the decreased content of syntaxin 1A in dST may be the part of the mechanism responsible for the radiation-induced hyperlocomotion.
Collapse
Affiliation(s)
- Viktor S Kokhan
- V.P. Serbsky National Medical Research Centre for Psychiatry and Narcology, Moscow, Russia.
| | - Kirill Chaprov
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry RAS, Chernogolovka, Russia
| | | | - Maxim S Nesterov
- Scientific Center for Biomedical Technologies of the Federal Biomedical Agency of Russia, settlement Svetlye Gory, Russia
| | - Vladimir A Pikalov
- Institute for High Energy Physics named by A.A. Logunov of NRC "Kurchatov Institute", Protvino, Russia
| |
Collapse
|
9
|
Chang HHV, Fesshaye AS, Tidmore A, Sanford LD, Britten RA. Sleep Fragmentation Results in Novel Set-shifting Decrements in GCR-exposed Male and Female Rats. Radiat Res 2025; 203:18-25. [PMID: 39576067 DOI: 10.1667/rade-24-00146.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 11/13/2024] [Indexed: 01/11/2025]
Abstract
The prolonged exposure to multiple spaceflight stressors during long-duration missions to the Moon and Mars will be challenging to the physical and mental health of the astronauts. Ground-based studies have reported that attentional set-shifting task (ATSET) performance is impaired after space radiation (SR) exposure. At certain times during deep-space missions, astronauts will likely have to contend with the combined impacts of SR and sleep perturbation. In rats, poor quality, fragmented sleep adversely impacts performance in multiple cognitive tasks, including the ATSET task. While both SR and sleep perturbations independently cause cognitive performance deficits, the incidence, severity and exact nature of those decrements following combined exposure to these flight stressors is largely unknown. This study established the impact that a single night of fragmented sleep has on ATSET performance in both male and female rats exposed to 10 cGy of galactic cosmic ray simulation (GCRsim). The GCRsim beam is a complex beam that mimics the mass and energy spectra of the SR particles that an astronaut will be exposed to within the spacecraft. Rats that had no obvious ATSET performance decrements when normally rested were subjected to fragmented sleep and their ATSET performance reassessed. Sleep fragmentation resulted in significant ATSET performance decrements in GCRsim-exposed rats, with specific performance decrements being observed in stages where attention or cue shifting is extensively used. Performance decrements in these stages are rarely observed after SR exposure. While both male and female rats exhibited latent sleep-related performance decrements, these were sex dependent, with male and female rats exhibiting different types of performance decrements (either reduced processing speed or task completion efficiency) in different stages of the ATSET task. This study suggests that SR-induced cognitive impairment may not be fully evident in normally rested rats, with an underestimation of both the incidence and nature of performance decrements that could occur when multiple space flight stressors are present. These data suggest that that there may be synergistic interactions between multiple space flight stressors that may not be easily predicted from their independent actions.
Collapse
Affiliation(s)
| | | | | | - Larry D Sanford
- Biomedical and Translational Sciences
- Center for Integrative Neuroscience and Inflammatory Diseases, Macon and Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, Virginia
| | - Richard A Britten
- Radiation Oncology
- Center for Integrative Neuroscience and Inflammatory Diseases, Macon and Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, Virginia
| |
Collapse
|
10
|
Skirzewski M, Skirzewski A. Beyond Earth's shield: The surprising way antioxidants could pave the road to Mars. J Neurochem 2025; 169:e16233. [PMID: 39387706 DOI: 10.1111/jnc.16233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 09/17/2024] [Accepted: 09/17/2024] [Indexed: 10/15/2024]
Abstract
Interplanetary travel poses serious risks because of Galactic cosmic radiations (GCRs). A recent study by Sanghee et al. revealed long-term cognitive impairments in female mice exposed to a 33-beam GCR simulator, highlighting persistent risks for astronauts. The study's use of touchscreen tasks, similar to human cognitive tests, enhances its relevance for space missions. Additionally, the antioxidant/anti-inflammatory compound CDDO-EA showed potential in mitigating these cognitive deficits. While offering critical insights into GCR effects, the study emphasizes the need for further research into protective strategies, including dietary interventions, to ensure astronaut safety on extended missions beyond Earth's protective shield.
Collapse
Affiliation(s)
- Miguel Skirzewski
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Aureliano Skirzewski
- Facultad de Ciencias, Instituto de Física, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
11
|
Krattli RP, Do AH, El-Khatib SM, Alikhani L, Markarian M, Vagadia AR, Usmani MT, Madan S, Baulch JE, Clark RJ, Woodruff TM, Tenner AJ, Acharya MM. C5aR1 inhibition alleviates cranial radiation-induced cognitive decline. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.02.601806. [PMID: 39005286 PMCID: PMC11245020 DOI: 10.1101/2024.07.02.601806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Cranial radiation therapy (RT) for brain cancers leads to an irreversible decline in cognitive function without an available remedy. Radiation-induced cognitive deficits (RICD) are a particularly pressing problem for the survivors of pediatric and low grade glioma (LGG) cancers who often live long post-RT lives. Radiation-induced elevated neuroinflammation and gliosis, triggered by the detrimental CNS complement cascade, lead to excessive synaptic and cognitive loss. Using intact and brain cancer-bearing mouse models, we now show that targeting anaphylatoxin complement C5a receptor (C5aR1) is neuroprotective against RICD. We used a genetic knockout, C5aR1 KO mouse, and a pharmacologic approach, employing the orally active, brain penetrant C5aR1 antagonist PMX205 to reverse RICD. Irradiated C5aR1 KO and WT mice receiving PMX205 showed significant neurocognitive improvements in object recognition memory and memory consolidation tasks. Inhibiting C5a/C5aR1 axis reduced microglial activation, astrogliosis, and synaptic loss in the irradiated brain. Importantly, C5aR1 blockage in two syngeneic, orthotopic glioblastoma-bearing mice protected against RICD without interfering with the therapeutic efficacy of RT to reduce tumor volume in vivo . PMX205 clinical trials with healthy individuals and amyotrophic lateral sclerosis (ALS) patients showed no toxicity, drug-related adverse events, or infections. Thus, C5aR1 inhibition is a translationally feasible approach to address RICD, an unmet medical need. SIGNIFICANCE Cranial radiotherapy for brain cancers activates CNS complement cascade, leading to cognitive decline. Ablation of the complement C5a/C5aR1 axis alleviates radiation-induced neuroinflammation, synaptic loss, and cognitive dysfunction, providing a novel tractable approach.
Collapse
|
12
|
Squire E, Lee HL, Jeong W, Lee S, Ravichandiran V, Limoli CL, Piomelli D, Parihar VK, Jung KM. Targeting dysfunctional endocannabinoid signaling in a mouse model of Gulf War illness. Neuropharmacology 2024; 261:110142. [PMID: 39241906 DOI: 10.1016/j.neuropharm.2024.110142] [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: 05/30/2024] [Revised: 07/30/2024] [Accepted: 09/02/2024] [Indexed: 09/09/2024]
Abstract
Gulf War Illness (GWI) is a chronic disorder characterized by a heterogeneous set of symptoms that include pain, fatigue, anxiety, and cognitive impairment. These are thought to stem from damage caused by exposure under unpredictable stress to toxic Gulf War (GW) chemicals, which include pesticides, nerve agents, and prophylactic drugs. We hypothesized that GWI pathogenesis might be rooted in long-lasting disruption of the endocannabinoid (ECB) system, a signaling complex that serves important protective functions in the brain. Using a mouse model of GWI, we found that tissue levels of the ECB messenger, anandamide, were significantly reduced in the brain of diseased mice, compared to healthy controls. In addition, transcription of the Faah gene, which encodes for fatty acid amide hydrolase (FAAH), the enzyme that deactivates anandamide, was significant elevated in prefrontal cortex of GWI mice and brain microglia. Behavioral deficits exhibited by these animals, including heightened anxiety-like and depression-like behaviors, and defective extinction of fearful memories, were corrected by administration of the FAAH inhibitor, URB597, which normalized brain anandamide levels. Furthermore, GWI mice displayed unexpected changes in the microglial transcriptome, implying persistent dampening of homeostatic surveillance genes and abnormal expression of pro-inflammatory genes upon immune stimulation. Together, these results suggest that exposure to GW chemicals produce a deficit in brain ECB signaling which is associated with persistent alterations in microglial function. Pharmacological normalization of anandamide-mediated ECB signaling may offer an effective therapeutic strategy for ameliorating GWI symptomology.
Collapse
Affiliation(s)
- Erica Squire
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, 92697, USA
| | - Hye-Lim Lee
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, 92697, USA
| | - Woojin Jeong
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, 92697, USA
| | - Sumin Lee
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, 92697, USA
| | - V Ravichandiran
- National Institute of Pharmaceutical Education and Research, Hajipur, Bihar, 844102, India
| | - Charles L Limoli
- Radiation Oncology, University of California, Irvine, CA 92697, USA
| | - Daniele Piomelli
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, 92697, USA; Department of Biological Chemistry, University of California, Irvine, CA, 92697, USA; Department of Pharmaceutical Sciences, University of California, Irvine, CA, 92697, USA
| | - Vipan Kumar Parihar
- National Institute of Pharmaceutical Education and Research, Hajipur, Bihar, 844102, India; Radiation Oncology, University of California, Irvine, CA 92697, USA.
| | - Kwang-Mook Jung
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, 92697, USA.
| |
Collapse
|
13
|
Britten RA, Fesshaye AS, Tidmore A, Tamgue EN, Alvarado-Arriaga PA. Different spectrum of space radiation induced cognitive impairments in radiation-naïve and adapted rats. LIFE SCIENCES IN SPACE RESEARCH 2024; 43:68-74. [PMID: 39521496 DOI: 10.1016/j.lssr.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/11/2024] [Accepted: 09/14/2024] [Indexed: 11/16/2024]
Abstract
NASA's decision to resume manned deep space mission, first to the Moon and then Mars, necessitated a detailed assessment of the potential health effects that astronauts may experience on long-duration missions. Multiple studies suggest that there may be significant space radiation (SR)-induced impairment of neurocognitive processes, including advanced executive functions. However, given the multitude of SR-induced changes in the CNS, it is possible that completely different SR-induced sequelae will be induced in previously exposed individuals. Thus, current risk estimates are likely to be pertinent only for the early stages of a deep space mission, and even then only for astronauts that have no previous experience in space. In this study, rats that maintained high attentional set shifting (ATSET) performance after an initial exposure to 10 cGy of SR (either 250 MeV/n He or GCRsim), were exposed to an additional dose of 10 cGy GCRsim and their ATSET performance reassessed. The re-irradiated rats exhibited significant impairment of ATSET performance, however, the performance decrements differed in two important aspects from those typically observed after single exposures. First, the decrements were manifested when the rats were required to perform set shifting, specifically in the IDR and EDS stages of the ATSET test. Secondly, the main performance decrement was in a loss of processing speed, which in the IDR stage resulted in the re-irradiated rats taking 2-fold more time to solve the problem than did Sham rats. The functional consequence of this decrement was that compared to Sham rats, 20 % fewer SR-exposed rats solved the IDS and EDR problems within 20 s. These data suggests that prior SR exposure alters nature of ATSET impairments from that observed in radiation-naïve individuals. Risk estimates derived from studies that use radiation naïve rats may thus not fully reflect the incidence and nature of ATSET performance deficits that could occur over the entire duration of a mission to Mars, or in astronauts who return to deep space on multiple occasions. It would thus be germane to conduct in-flight monitoring for cognitive performance decrements observed in both radiation naïve and exposed rats during the mission, and ensure that the crew has sufficient overlapping skill sets to minimize the operational impact of these additional cognitive impairments.
Collapse
Affiliation(s)
- Richard A Britten
- Radiation Oncology, USA; Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Macon and Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA. 23507, USA.
| | | | | | | | | |
Collapse
|
14
|
McGhee HG, Totani R, Plekan O, Coreno M, de Simone M, Mumtaz A, Singh S, Schroeder BC, Curchod BFE, Ingle RA. Core and valence photoelectron spectroscopy of a series of substituted disulfides. J Chem Phys 2024; 161:134303. [PMID: 39356064 DOI: 10.1063/5.0231178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 09/06/2024] [Indexed: 10/03/2024] Open
Abstract
The valence and core photoelectron spectra of three substituted disulfide systems, α-lipoic acid, trans-4,5-dihydroxy-1,2-dithiane, and di-Boc-cystamine, are presented alongside detailed theoretical analysis based on equation-of-motion coupled-cluster singles doubles for ionization potentials and the nuclear ensemble approach. A comparison of the linear and five- and six-membered ring cyclic structures reveals that the energetic separation of the non-bonding sulfur orbitals can be used to calculate a reliable estimate of the C-S-S-C dihedral angle, even for substituted disulfides, and that the sulfur 2p, oxygen 1s, and valence band photoelectron spectra are a useful site-specific probe of hydrogen bonding.
Collapse
Affiliation(s)
- H G McGhee
- Department of Chemistry, 20 Gordon Street, London WC1H 0AJ, United Kingdom
| | - R Totani
- ISM-CNR, Istituto di Struttura dei Materiali, LD2 Unit, 34149 Trieste, Italy
- Elettra-Sincrotrone Trieste, I-34149 Basovizza, Trieste, Italy
| | - O Plekan
- ISM-CNR, Istituto di Struttura dei Materiali, LD2 Unit, 34149 Trieste, Italy
| | - M Coreno
- ISM-CNR, Istituto di Struttura dei Materiali, LD2 Unit, 34149 Trieste, Italy
| | - M de Simone
- IOM-CNR, Istituto Officina dei Materiali, 34149 Trieste, Italy
| | - A Mumtaz
- Department of Chemistry, 20 Gordon Street, London WC1H 0AJ, United Kingdom
| | - S Singh
- Department of Chemistry, 20 Gordon Street, London WC1H 0AJ, United Kingdom
| | - B C Schroeder
- Department of Chemistry, 20 Gordon Street, London WC1H 0AJ, United Kingdom
| | - B F E Curchod
- Centre for Computational Chemistry, School of Chemistry, University of Bristol, Bristol BS8 1TS, United Kingdom
| | - R A Ingle
- Department of Chemistry, 20 Gordon Street, London WC1H 0AJ, United Kingdom
| |
Collapse
|
15
|
Sleiman A, Miller KB, Flores D, Kuan J, Altwasser K, Smith BJ, Kozbenko T, Hocking R, Wood SJ, Huff J, Adam-Guillermin C, Hamada N, Yauk C, Wilkins R, Chauhan V. AOP report: Development of an adverse outcome pathway for deposition of energy leading to learning and memory impairment. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2024; 65 Suppl 3:57-84. [PMID: 39228295 DOI: 10.1002/em.22622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 09/05/2024]
Abstract
Understanding radiation-induced non-cancer effects on the central nervous system (CNS) is essential for the risk assessment of medical (e.g., radiotherapy) and occupational (e.g., nuclear workers and astronauts) exposures. Herein, the adverse outcome pathway (AOP) approach was used to consolidate relevant studies in the area of cognitive decline for identification of research gaps, countermeasure development, and for eventual use in risk assessments. AOPs are an analytical construct describing critical events to an adverse outcome (AO) in a simplified form beginning with a molecular initiating event (MIE). An AOP was constructed utilizing mechanistic information to build empirical support for the key event relationships (KERs) between the MIE of deposition of energy to the AO of learning and memory impairment through multiple key events (KEs). The evidence for the AOP was acquired through a documented scoping review of the literature. In this AOP, the MIE is connected to the AO via six KEs: increased oxidative stress, increased deoxyribonucleic acid (DNA) strand breaks, altered stress response signaling, tissue resident cell activation, increased pro-inflammatory mediators, and abnormal neural remodeling that encompasses atypical structural and functional alterations of neural cells and surrounding environment. Deposition of energy directly leads to oxidative stress, increased DNA strand breaks, an increase of pro-inflammatory mediators and tissue resident cell activation. These KEs, which are themselves interconnected, can lead to abnormal neural remodeling impacting learning and memory processes. Identified knowledge gaps include improving quantitative understanding of the AOP across several KERs and additional testing of proposed modulating factors through experimental work. Broadly, it is envisioned that the outcome of these efforts could be extended to other cognitive disorders and complement ongoing work by international radiation governing bodies in their review of the system of radiological protection.
Collapse
Affiliation(s)
- Ahmad Sleiman
- Institut de Radioprotection et de Sûreté Nucléaire, St. Paul Lez Durance, Provence, France
| | - Kathleen B Miller
- Department of Health and Exercise Science, Morrison College Family of Health, University of St. Thomas, Saint Paul, Minnesota, USA
| | - Danicia Flores
- Consumer and Clinical Radiation Protection Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Ontario, Canada
| | - Jaqueline Kuan
- Consumer and Clinical Radiation Protection Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Ontario, Canada
| | - Kaitlyn Altwasser
- Consumer and Clinical Radiation Protection Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Ontario, Canada
| | - Benjamin J Smith
- Consumer and Clinical Radiation Protection Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Ontario, Canada
| | - Tatiana Kozbenko
- Consumer and Clinical Radiation Protection Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Ontario, Canada
| | - Robyn Hocking
- Consumer and Clinical Radiation Protection Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Ontario, Canada
| | | | - Janice Huff
- NASA Langley Research Center, Hampton, Virginia, USA
| | | | - Nobuyuki Hamada
- Biology and Environmental Chemistry Division, Sustainable System Research Laboratory, Central Research Institute of Electric Power Industry (CRIEPI), Chiba, Japan
| | - Carole Yauk
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Ruth Wilkins
- Consumer and Clinical Radiation Protection Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Ontario, Canada
| | - Vinita Chauhan
- Consumer and Clinical Radiation Protection Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Ontario, Canada
| |
Collapse
|
16
|
Wang E, Shuryak I, Brenner DJ. A competing risks machine learning study of neutron dose, fractionation, age, and sex effects on mortality in 21,000 mice. Sci Rep 2024; 14:17974. [PMID: 39095647 PMCID: PMC11297256 DOI: 10.1038/s41598-024-68717-9] [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: 03/25/2024] [Accepted: 07/25/2024] [Indexed: 08/04/2024] Open
Abstract
This study explores the impact of densely-ionizing radiation on non-cancer and cancer diseases, focusing on dose, fractionation, age, and sex effects. Using historical mortality data from approximately 21,000 mice exposed to fission neutrons, we employed random survival forest (RSF), a powerful machine learning algorithm accommodating nonlinear dependencies and interactions, treating cancer and non-cancer outcomes as competing risks. Unlike traditional parametric models, RSF avoids strict assumptions and captures complex data relationships through decision tree ensembles. SHAP (SHapley Additive exPlanations) values and variable importance scores were employed for interpretation. The findings revealed clear dose-response trends, with cancer being the predominant cause of mortality. SHAP value dose-response shapes differed, showing saturation for cancer hazard at high doses (> 2 Gy) and a more linear pattern at lower doses. Non-cancer responses remained more linear throughout the entire dose range. There was a potential inverse dose rate effect for cancer, while the evidence for non-cancer was less conclusive. Sex and age effects were less pronounced. This investigation, utilizing machine learning, enhances our understanding of the patterns of non-cancer and cancer mortality induced by densely-ionizing radiations, emphasizing the importance of such approaches in radiation research, including space travel and radioprotection.
Collapse
Affiliation(s)
- Eric Wang
- Center for Radiological Research, Columbia University Irving Medical Center, 630 West 168th street, VC-11, New York, NY, 10032, USA.
| | - Igor Shuryak
- Center for Radiological Research, Columbia University Irving Medical Center, 630 West 168th street, VC-11, New York, NY, 10032, USA
| | - David J Brenner
- Center for Radiological Research, Columbia University Irving Medical Center, 630 West 168th street, VC-11, New York, NY, 10032, USA
| |
Collapse
|
17
|
Dickstein DL, Zhang R, Ru N, Vozenin MC, Perry BC, Wang J, Baulch J, Acharya MM, Limoli CL. Structural plasticity of pyramidal cell neurons measured after FLASH and conventional dose-rate irradiation. RESEARCH SQUARE 2024:rs.3.rs-4656938. [PMID: 39108471 PMCID: PMC11302692 DOI: 10.21203/rs.3.rs-4656938/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Evidence shows that ultra-high dose-rate FLASH-radiotherapy (FLASH-RT) protects against normal tissue complications and functional decrements in the irradiated brain. Past work has shown that radiation-induced cognitive impairment, neuroinflammation and reduced structural complexity of granule cell neurons were not observed to the same extent after FLASH-RT (> MGy/s) compared to conventional dose-rate (CONV, 0.1 Gy/s) delivery. To explore the sensitivity of different neuronal populations to cranial irradiation and dose-rate modulation, hippocampal CA1 and medial prefrontal cortex (PFC) pyramidal neurons were analyzed by electron and confocal microscopy. Neuron ultrastructural analyses by electron microscopy after 10 Gy FLASH- or CONV-RT exposures indicated that irradiation had little impact on dendritic complexity and synapse density in the CA1, but did increase length and head diameter of smaller non-perforated synapses. Similarly, irradiation caused no change in PFC prelimbic/infralimbic axospinous synapse density, but reductions in non-perforated synapse diameters. While irradiation resulted in thinner myelin sheaths compared to controls, none of these metrics were dose-rate sensitive. Analysis of fluorescently labeled CA1 neurons revealed no radiation-induced or dose-rate-dependent changes in overall dendritic complexity or spine density, in contrast to our past analysis of granule cell neurons. Super-resolution confocal microscopy following a clinical dosing paradigm (3×10Gy) showed significant reductions in excitatory vesicular glutamate transporter 1 and inhibitory vesicular GABA transporter puncta density within the CA1 that were largely dose-rate independent. Collectively, these data reveal that, compared to granule cell neurons, CA1 and mPFC neurons are more radioresistant irrespective of radiation dose-rate.
Collapse
Affiliation(s)
| | | | - Ning Ru
- University of California, Irvine School of Medicine
| | | | | | - Juan Wang
- Uniformed Services University of Health Sciences
| | - Janet Baulch
- University of California, Irvine School of Medicine
| | | | | |
Collapse
|
18
|
Impey S, Raber J. Irradiation and Alterations in Hippocampal DNA Methylation. EPIGENOMES 2024; 8:27. [PMID: 39051185 PMCID: PMC11270359 DOI: 10.3390/epigenomes8030027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 06/11/2024] [Accepted: 07/02/2024] [Indexed: 07/27/2024] Open
Abstract
The response of the brain to radiation is important for cancer patients receiving whole or partial brain irradiation or total body irradiation, those exposed to irradiation as part of a nuclear accident or a nuclear war or terrorism event, and for astronauts during and following space missions. The mechanisms mediating the effects of irradiation on the hippocampus might be associated with alterations in hippocampal DNA methylation. Changes in cytosine methylation involving the addition of a methyl group to cytosine (5 mC) and especially those involving the addition of a hydroxy group to 5 mC (hydroxymethylcytosine or 5 hmC) play a key role in regulating the expression of genes required for hippocampal function. In this review article, we will discuss the effects of radiation on hippocampal DNA methylation and whether these effects are associated with hippocampus-dependent cognitive measures and molecular measures in the hippocampus involved in cognitive measures. We will also discuss whether the radiation-induced changes in hippocampal DNA methylation show an overlap across different doses of heavy ion irradiation and across irradiation with different ions. We will also discuss whether the DNA methylation changes show a tissue-dependent response.
Collapse
Affiliation(s)
- Soren Impey
- Dow Neurobiology Laboratories, Legacy Research Institute Legacy Health Systems, 1225 NE 2nd Ave, Portland, OR 97232, USA
- Departments of Behavioral Neuroscience, Neurology, and Radiation Medicine, Division of Neuroscience, ONPRC, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Jacob Raber
- Departments of Behavioral Neuroscience, Neurology, and Radiation Medicine, Division of Neuroscience, ONPRC, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| |
Collapse
|
19
|
Hinkle JJ, Olschowka JA, Williams JP, O'Banion MK. Pharmacologic Manipulation of Complement Receptor 3 Prevents Dendritic Spine Loss and Cognitive Impairment After Acute Cranial Radiation. Int J Radiat Oncol Biol Phys 2024; 119:912-923. [PMID: 38142839 DOI: 10.1016/j.ijrobp.2023.12.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 12/08/2023] [Accepted: 12/15/2023] [Indexed: 12/26/2023]
Abstract
PURPOSE Cranial irradiation induces healthy tissue damage that can lead to neurocognitive complications, negatively affecting patient quality of life. One damage indicator associated with cognitive impairment is loss of neuronal spine density. We previously demonstrated that irradiation-mediated spine loss is microglial complement receptor 3 (CR3) and sex dependent. We hypothesized that these changes are associated with late-delayed cognitive deficits and amenable to pharmacologic intervention. METHODS AND MATERIALS Our model of cranial irradiation (acute, 10 Gy gamma) used male and female CR3-wild type and CR3-deficient Thy-1 YFP mice of C57BL/6 background. Forty-five days after irradiation and behavioral testing, we quantified spine density and markers of microglial reactivity in the hippocampal dentate gyrus. In a separate experiment, male Thy-1 YFP C57BL/6 mice were treated with leukadherin-1, a modulator of CR3 function. RESULTS We found that male mice demonstrate irradiation-mediated spine loss and cognitive deficits but that female and CR3 knockout mice do not. These changes were associated with greater reactivity of microglia in male mice. Pharmacologic manipulation of CR3 with LA1 prevented spine loss and cognitive deficits in irradiated male mice. CONCLUSIONS This work improves our understanding of irradiation-mediated mechanisms and sex dependent responses and may help identify novel therapeutics to reduce irradiation-induced cognitive decline and improve patient quality of life.
Collapse
Affiliation(s)
- Joshua J Hinkle
- Department of Neuroscience and Del Monte Neuroscience Institute
| | | | | | - M Kerry O'Banion
- Department of Neuroscience and Del Monte Neuroscience Institute; Department of Neurology, University of Rochester School of Medicine and Dentistry, Rochester, New York.
| |
Collapse
|
20
|
Kokhan VS, Pikalov VA, Chaprov K, Gulyaev MV. Combined Ionizing Radiation Exposure by Gamma Rays and Carbon-12 Nuclei Increases Neurotrophic Factor Content and Prevents Age-Associated Decreases in the Volume of the Sensorimotor Cortex in Rats. Int J Mol Sci 2024; 25:6725. [PMID: 38928431 PMCID: PMC11203503 DOI: 10.3390/ijms25126725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/08/2024] [Accepted: 06/16/2024] [Indexed: 06/28/2024] Open
Abstract
In orbital and ground-based experiments, it has been demonstrated that ionizing radiation (IR) can stimulate the locomotor and exploratory activity of rodents, but the underlying mechanism of this phenomenon remains undisclosed. Here, we studied the effect of combined IR (0.4 Gy γ-rays and 0.14 Gy carbon-12 nuclei) on the locomotor and exploratory activity of rats, and assessed the sensorimotor cortex volume by magnetic resonance imaging-based morphometry at 1 week and 7 months post-irradiation. The sensorimotor cortex tissues were processed to determine whether the behavioral and morphologic effects were associated with changes in neurotrophin content. The irradiated rats were characterized by increased locomotor and exploratory activity, as well as novelty-seeking behavior, at 3 days post-irradiation. At the same time, only unirradiated rats experienced a significant decrease in the sensorimotor cortex volume at 7 months. While there were no significant differences at 1 week, at 7 months, the irradiated rats were characterized by higher neurotrophin-3 and neurotrophin-4 content in the sensorimotor cortex. Thus, IR prevents the age-associated decrease in the sensorimotor cortex volume, which is associated with neurotrophic and neurogenic changes. Meanwhile, IR-induced increases in locomotor activity may be the cause of the observed changes.
Collapse
Affiliation(s)
- Viktor S. Kokhan
- V.P. Serbsky National Medical Research Centre for Psychiatry and Narcology, 119034 Moscow, Russia
| | - Vladimir A. Pikalov
- Institute for High Energy Physics Named by A.A. Logunov of NRC “Kurchatov Institute”, 142281 Protvino, Russia;
| | - Kirill Chaprov
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences, 142432 Chernogolovka, Russia;
| | - Mikhail V. Gulyaev
- Faculty of Medicine, M.V. Lomonosov Moscow State University, 119991 Moscow, Russia;
| |
Collapse
|
21
|
Masarapu Y, Cekanaviciute E, Andrusivova Z, Westholm JO, Björklund Å, Fallegger R, Badia-I-Mompel P, Boyko V, Vasisht S, Saravia-Butler A, Gebre S, Lázár E, Graziano M, Frapard S, Hinshaw RG, Bergmann O, Taylor DM, Wallace DC, Sylvén C, Meletis K, Saez-Rodriguez J, Galazka JM, Costes SV, Giacomello S. Spatially resolved multiomics on the neuronal effects induced by spaceflight in mice. Nat Commun 2024; 15:4778. [PMID: 38862479 PMCID: PMC11166911 DOI: 10.1038/s41467-024-48916-8] [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: 05/23/2023] [Accepted: 05/17/2024] [Indexed: 06/13/2024] Open
Abstract
Impairment of the central nervous system (CNS) poses a significant health risk for astronauts during long-duration space missions. In this study, we employed an innovative approach by integrating single-cell multiomics (transcriptomics and chromatin accessibility) with spatial transcriptomics to elucidate the impact of spaceflight on the mouse brain in female mice. Our comparative analysis between ground control and spaceflight-exposed animals revealed significant alterations in essential brain processes including neurogenesis, synaptogenesis and synaptic transmission, particularly affecting the cortex, hippocampus, striatum and neuroendocrine structures. Additionally, we observed astrocyte activation and signs of immune dysfunction. At the pathway level, some spaceflight-induced changes in the brain exhibit similarities with neurodegenerative disorders, marked by oxidative stress and protein misfolding. Our integrated spatial multiomics approach serves as a stepping stone towards understanding spaceflight-induced CNS impairments at the level of individual brain regions and cell types, and provides a basis for comparison in future spaceflight studies. For broader scientific impact, all datasets from this study are available through an interactive data portal, as well as the National Aeronautics and Space Administration (NASA) Open Science Data Repository (OSDR).
Collapse
Affiliation(s)
- Yuvarani Masarapu
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Egle Cekanaviciute
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, Mountain View, CA, 94035, USA
| | - Zaneta Andrusivova
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Jakub O Westholm
- National Bioinformatics Infrastructure Sweden, Department of Biochemistry and Biophysics, Stockholm University, Science for Life Laboratory, Stockholm, Sweden
| | - Åsa Björklund
- Department of Cell and Molecular Biology, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Robin Fallegger
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Bioquant, Heidelberg, Germany
| | - Pau Badia-I-Mompel
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Bioquant, Heidelberg, Germany
- GSK, Cellzome, Heidelberg, Germany
| | - Valery Boyko
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, Mountain View, CA, 94035, USA
- Bionetics, Yorktown, VA, USA
| | - Shubha Vasisht
- Department of Biomedical and Health Informatics, The Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
| | - Amanda Saravia-Butler
- KBR, Space Biosciences Division, NASA Ames Research Center, Moffett Field, Mountain View, CA, 94035, USA
| | - Samrawit Gebre
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, Mountain View, CA, 94035, USA
| | - Enikő Lázár
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Marta Graziano
- Department of Neuroscience, Karolinska Institutet, Biomedicum, Solna, Sweden
| | - Solène Frapard
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Robert G Hinshaw
- NASA Postdoctoral Program - Oak Ridge Associated Universities, NASA Ames Research Center, Moffett Field, Mountain View, CA, 94035, USA
| | - Olaf Bergmann
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
- Pharmacology and Toxicology, Department of Pharmacology and Toxicology University Medical Center Goettingen, Goettingen, Germany
| | - Deanne M Taylor
- Department of Biomedical and Health Informatics, The Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
- Department of Pediatrics, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Douglas C Wallace
- Center for Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia and Department of Pediatrics, Division of Human Genetics, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Christer Sylvén
- Department of Medicine, Karolinska Institute, Huddinge, Sweden
| | | | - Julio Saez-Rodriguez
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Bioquant, Heidelberg, Germany
| | - Jonathan M Galazka
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, Mountain View, CA, 94035, USA
| | - Sylvain V Costes
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, Mountain View, CA, 94035, USA.
| | - Stefania Giacomello
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden.
| |
Collapse
|
22
|
Bevelacqua JJ, Mortazavi SAR, Mortazavi SMJ, Welsh J. Professor John Roderick Cameron's Influence on Radiation Safety in Terrestrial and Space Environments. J Biomed Phys Eng 2024; 14:319-322. [PMID: 39027714 PMCID: PMC11252544 DOI: 10.31661/jbpe.v0i0.2405-1767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 05/26/2024] [Indexed: 07/20/2024]
Abstract
Professor John Roderick Cameron (1922-2005) stands out as a trailblazer in the field of medical physics, whose innovative work has deeply influenced radiation protection and the broader medical radiation field through sound technical judgment and insight. Best known for inventing the bone densitometry device, his pioneering efforts have reshaped modern medical practices far beyond his initial breakthroughs. Cameron's explorations extended into the realms of space biomedical science and models of terrestrial radiation, areas where his insights continue to resonate today. As the Emeritus Professor of Medical Physics at the University of Wisconsin-Madison and a founding member of the American Association of Physicists in Medicine, Cameron laid crucial groundwork for safety standards in environments with high natural radiation levels. His leadership was instrumental in advancing thermoluminescence dosimetry, radiation measurement, and image quality assurance, driving progress in both academia and clinical practices. Moreover, through establishing Medical Physics Publishing, Cameron played a pivotal role in spreading vital research and educational materials across the fields of health physics and medical physics. This commentary reflects on Cameron's far-reaching contributions, highlighting his critical work in space radiation research and terrestrial radiation models-key to the future of interplanetary travel and potential human settlement on planets like Mars. His research in areas of high background radiation, like Ramsar, Iran, has been fundamental in developing strategies for biological protection in space, which are essential for ensuring astronaut safety during long-duration space missions. We honor Professor Cameron's profound legacy, celebrating his visionary spirit and the lasting impact of his contributions on generations of scientists in radiation science.
Collapse
Affiliation(s)
| | | | - Seyed Mohammad Javad Mortazavi
- Ionizing and Non-Ionizing Radiation Protection Research Center (INIRPRC), Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Medical Physics and Engineering, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - James Welsh
- Department of Radiation Oncology, Edward Hines Jr VA Hospital, Hines, IL, United States
- Department of Radiation Oncology, Stritch School of Medicine, Loyola University, Chicago, IL, United States
| |
Collapse
|
23
|
Cahoon DS, Fisher DR, Rabin BM, Lamon-Fava S, Wu D, Zheng T, Shukitt-Hale B. Galactic Cosmic Ray Particle Exposure Does Not Increase Protein Levels of Inflammation or Oxidative Stress Markers in Rat Microglial Cells In Vitro. Int J Mol Sci 2024; 25:5923. [PMID: 38892109 PMCID: PMC11172496 DOI: 10.3390/ijms25115923] [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: 03/14/2024] [Revised: 05/22/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Astronauts on exploratory missions will be exposed to galactic cosmic rays (GCR), which can induce neuroinflammation and oxidative stress (OS) and may increase the risk of neurodegenerative disease. As key regulators of inflammation and OS in the CNS, microglial cells may be involved in GCR-induced deficits, and therefore could be a target for neuroprotection. This study assessed the effects of exposure to helium (4He) and iron (56Fe) particles on inflammation and OS in microglia in vitro, to establish a model for testing countermeasure efficacy. Rat microglia were exposed to a single dose of 20 cGy (300 MeV/n) 4He or 2 Gy 56Fe (600 MeV/n), while the control cells were not exposed (0 cGy). Immediately following irradiation, fresh media was applied to the cells, and biomarkers of inflammation (cyclooxygenase-2 [COX-2], nitric oxide synthase [iNOS], phosphorylated IκB-α [pIκB-α], tumor necrosis factor-α [TNFα], and nitrite [NO2-]) and OS (NADPH oxidase [NOX2]) were assessed 24 h later using standard immunochemical techniques. Results showed that radiation did not increase levels of NO2- or protein levels of COX-2, iNOS, pIκB-α, TNFα, or NOX2 compared to non-irradiated control conditions in microglial cells (p > 0.05). Therefore, microglia in isolation may not be the primary cause of neuroinflammation and OS following exposures to helium or iron GCR particles.
Collapse
Affiliation(s)
- Danielle S. Cahoon
- USDA-ARS, Human Nutrition Research Center on Aging at Tufts University, Boston, MA 02111, USA; (D.S.C.); (D.R.F.); (S.L.-F.); (D.W.); (T.Z.)
| | - Derek R. Fisher
- USDA-ARS, Human Nutrition Research Center on Aging at Tufts University, Boston, MA 02111, USA; (D.S.C.); (D.R.F.); (S.L.-F.); (D.W.); (T.Z.)
| | - Bernard M. Rabin
- Department of Psychology, University of Maryland, Baltimore County (UMBC), Baltimore, MD 21250, USA;
| | - Stefania Lamon-Fava
- USDA-ARS, Human Nutrition Research Center on Aging at Tufts University, Boston, MA 02111, USA; (D.S.C.); (D.R.F.); (S.L.-F.); (D.W.); (T.Z.)
| | - Dayong Wu
- USDA-ARS, Human Nutrition Research Center on Aging at Tufts University, Boston, MA 02111, USA; (D.S.C.); (D.R.F.); (S.L.-F.); (D.W.); (T.Z.)
| | - Tong Zheng
- USDA-ARS, Human Nutrition Research Center on Aging at Tufts University, Boston, MA 02111, USA; (D.S.C.); (D.R.F.); (S.L.-F.); (D.W.); (T.Z.)
| | - Barbara Shukitt-Hale
- USDA-ARS, Human Nutrition Research Center on Aging at Tufts University, Boston, MA 02111, USA; (D.S.C.); (D.R.F.); (S.L.-F.); (D.W.); (T.Z.)
| |
Collapse
|
24
|
Adkins AM, Luyo ZNM, Gibbs AJ, Boden AF, Heerbrandt RS, Gotthold JD, Britten RA, Wellman LL, Sanford LD. Alterations in Blood-Brain Barrier Integrity and Lateral Ventricle Differ in Rats Exposed to Space Radiation and Social Isolation. Life (Basel) 2024; 14:636. [PMID: 38792656 PMCID: PMC11122575 DOI: 10.3390/life14050636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/03/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
The proposed Mars missions will expose astronauts to long durations of social isolation (SI) and space radiation (SR). These stressors have been shown to alter the brain's macrostructure and microenvironment, including the blood-brain barrier (BBB). Breakdown of the BBB is linked to impaired executive functions and physical deficits, including sensorimotor and neurocognitive impairments. However, the precise mechanisms mediating these effects remain unknown. Additionally, the synergistic effects of combined exposure to SI and SR on the structural integrity of the BBB and brain remain unknown. We assessed the BBB integrity and morphology in the brains of male rats exposed to ground-based analogs of SI and SR. The rats exposed to SR had enlarged lateral ventricles and increased BBB damage associated with a loss of astrocytes and an increased number of leaky vessels. Many deficits observed in SR-treated animals were attenuated by dual exposure to SI (DFS). SI alone did not show BBB damage but did show differences in astrocyte morphology compared to the Controls. Thus, determining how single and combined inflight stressors modulate CNS structural integrity is crucial to fully understand the multiple pathways that could impact astronaut performance and health, including the alterations to the CNS structures and cell viability observed in this study.
Collapse
Affiliation(s)
- Austin M. Adkins
- Sleep Research Laboratory, Center for Integrative Neuroscience and Inflammatory Diseases, Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (A.M.A.); (Z.N.M.L.); (A.F.B.); (R.S.H.); (J.D.G.); (L.L.W.)
| | - Zachary N. M. Luyo
- Sleep Research Laboratory, Center for Integrative Neuroscience and Inflammatory Diseases, Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (A.M.A.); (Z.N.M.L.); (A.F.B.); (R.S.H.); (J.D.G.); (L.L.W.)
| | - Alayna J. Gibbs
- Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA;
| | - Alea F. Boden
- Sleep Research Laboratory, Center for Integrative Neuroscience and Inflammatory Diseases, Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (A.M.A.); (Z.N.M.L.); (A.F.B.); (R.S.H.); (J.D.G.); (L.L.W.)
| | - Riley S. Heerbrandt
- Sleep Research Laboratory, Center for Integrative Neuroscience and Inflammatory Diseases, Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (A.M.A.); (Z.N.M.L.); (A.F.B.); (R.S.H.); (J.D.G.); (L.L.W.)
| | - Justin D. Gotthold
- Sleep Research Laboratory, Center for Integrative Neuroscience and Inflammatory Diseases, Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (A.M.A.); (Z.N.M.L.); (A.F.B.); (R.S.H.); (J.D.G.); (L.L.W.)
| | - Richard A. Britten
- Center for Integrative Neuroscience and Inflammatory Diseases, Radiation Oncology, Eastern Virginia Medical School, Norfolk, VA 23507, USA;
| | - Laurie L. Wellman
- Sleep Research Laboratory, Center for Integrative Neuroscience and Inflammatory Diseases, Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (A.M.A.); (Z.N.M.L.); (A.F.B.); (R.S.H.); (J.D.G.); (L.L.W.)
| | - Larry D. Sanford
- Sleep Research Laboratory, Center for Integrative Neuroscience and Inflammatory Diseases, Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (A.M.A.); (Z.N.M.L.); (A.F.B.); (R.S.H.); (J.D.G.); (L.L.W.)
| |
Collapse
|
25
|
Adkins AM, Colby EM, Boden AF, Gotthold JD, Harris RD, Britten RA, Wellman LL, Sanford LD. Effects of social isolation and galactic cosmic radiation on fine motor skills and behavioral performance. LIFE SCIENCES IN SPACE RESEARCH 2024; 41:74-79. [PMID: 38670655 DOI: 10.1016/j.lssr.2024.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/19/2024] [Accepted: 01/25/2024] [Indexed: 04/28/2024]
Abstract
Future NASA missions will require astronauts to travel farther and spend longer durations in space than ever before. This will also expose astronauts to longer periods of several physical and psychological challenges, including exposure to space radiation (SR) and periods of social isolation (SI), which could have unknown negative effects on physical and mental health. Each also has the potential to negatively impact sleep which can reduce the ability to cope with stressful experiences and lead to sensorimotor, neurocognitive, and physical deficits. The effects of SI and SR on gross motor performance has been shown to vary, and depend on, individual differences in stress resilience and vulnerability based on our established animal model in which stress produces different effects on sleep. In this study, the impact that SI and SR, either alone or together, had on fine motor skill performance (bilateral tactile adhesive removal task (BTAR)) was assessed in male rats. We also examined emotional, exploratory, and other off-task behavioral responses during testing and assessed whether sensorimotor performance and emotion varied with individual differences in resilience and vulnerability. BTAR task performance was differentially impacted by SI and SR, and were further influenced by the stress resilience/vulnerability phenotype of the rats. These findings further demonstrate that identifying individual responses to stressors that can impact sensorimotor ability and behavior necessary to perform mission-related tasks will be of particular importance for astronauts and future missions. Should similar effects occur in humans, there may be considerable inter-individual variability in the impact that inflight stressors have on astronauts and their ability to perform mission-related tasks.
Collapse
Affiliation(s)
- Austin M Adkins
- Sleep Research Laboratory, Eastern Virginia Medical School, Norfolk, VA, USA; Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA, USA; Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Emily M Colby
- Sleep Research Laboratory, Eastern Virginia Medical School, Norfolk, VA, USA; Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA, USA; Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Alea F Boden
- Sleep Research Laboratory, Eastern Virginia Medical School, Norfolk, VA, USA; Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA, USA; Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Justin D Gotthold
- Sleep Research Laboratory, Eastern Virginia Medical School, Norfolk, VA, USA; Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA, USA; Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Ryan D Harris
- Sleep Research Laboratory, Eastern Virginia Medical School, Norfolk, VA, USA; Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA, USA; Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Richard A Britten
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA, USA; Radiation Oncology, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Laurie L Wellman
- Sleep Research Laboratory, Eastern Virginia Medical School, Norfolk, VA, USA; Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA, USA; Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Larry D Sanford
- Sleep Research Laboratory, Eastern Virginia Medical School, Norfolk, VA, USA; Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA, USA; Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA, USA.
| |
Collapse
|
26
|
Iacono D, Hatch K, Murphy EK, Post J, Cole RN, Perl DP, Day RM. Proteomic changes in the hippocampus of large mammals after total-body low dose radiation. PLoS One 2024; 19:e0296903. [PMID: 38427613 PMCID: PMC10906861 DOI: 10.1371/journal.pone.0296903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 12/19/2023] [Indexed: 03/03/2024] Open
Abstract
There is a growing interest in low dose radiation (LDR) to counteract neurodegeneration. However, LDR effects on normal brain have not been completely explored yet. Recent analyses showed that LDR exposure to normal brain tissue causes expression level changes of different proteins including neurodegeneration-associated proteins. We assessed the proteomic changes occurring in radiated vs. sham normal swine brains. Due to its involvement in various neurodegenerative processes, including those associated with cognitive changes after high dose radiation exposure, we focused on the hippocampus first. We observed significant proteomic changes in the hippocampus of radiated vs. sham swine after LDR (1.79Gy). Mass spectrometry results showed 190 up-regulated and 120 down-regulated proteins after LDR. Western blotting analyses confirmed increased levels of TPM1, TPM4, PCP4 and NPY (all proteins decreased in various neurodegenerative processes, with NPY and PCP4 known to be neuroprotective) in radiated vs. sham swine. These data support the use of LDR as a potential beneficial tool to interfere with neurodegenerative processes and perhaps other brain-related disorders, including behavioral disorders.
Collapse
Affiliation(s)
- Diego Iacono
- DoD/USU Brain Tissue Repository & Neuropathology Program, Uniformed Services University (USU), Bethesda, Maryland, United States of America
- Department of Neurology, F. Edward Hébert School of Medicine, Uniformed Services University (USU), Bethesda, Maryland, United States of America
- Department of Pathology, F. Edward Hébert School of Medicine, Uniformed Services University (USU), Bethesda, Maryland, United States of America
- Neuroscience Program, Department of Anatomy, Physiology and Genetics (APG), F. Edward Hébert School of Medicine, Uniformed Services University (USU), Bethesda, Maryland, United States of America
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc. (HJF), Bethesda, Maryland, United States of America
- Neurodegeneration Disorders Clinic, National Institute of Neurological Disorders and Stroke, NINDS, NIH, Bethesda, Maryland, United States of America
| | - Kathleen Hatch
- DoD/USU Brain Tissue Repository & Neuropathology Program, Uniformed Services University (USU), Bethesda, Maryland, United States of America
- Department of Pathology, F. Edward Hébert School of Medicine, Uniformed Services University (USU), Bethesda, Maryland, United States of America
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc. (HJF), Bethesda, Maryland, United States of America
| | - Erin K. Murphy
- DoD/USU Brain Tissue Repository & Neuropathology Program, Uniformed Services University (USU), Bethesda, Maryland, United States of America
- Department of Pathology, F. Edward Hébert School of Medicine, Uniformed Services University (USU), Bethesda, Maryland, United States of America
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc. (HJF), Bethesda, Maryland, United States of America
| | - Jeremy Post
- Mass Spectrometry and Proteomics, Department of Biological Chemistry, Johns Hopkins University, School of Medicine, Baltimore, Maryland, United States of America
| | - Robert N. Cole
- Mass Spectrometry and Proteomics, Department of Biological Chemistry, Johns Hopkins University, School of Medicine, Baltimore, Maryland, United States of America
| | - Daniel P. Perl
- DoD/USU Brain Tissue Repository & Neuropathology Program, Uniformed Services University (USU), Bethesda, Maryland, United States of America
- Department of Pathology, F. Edward Hébert School of Medicine, Uniformed Services University (USU), Bethesda, Maryland, United States of America
| | - Regina M. Day
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University (USU), Bethesda, Maryland, United States of America
| |
Collapse
|
27
|
Chen Z, Li Y, Rasheed M, Wang H, Lei R, Zhao T, Deng Y, Ma H. Corrigendum: Altered expression of inflammation-associated molecules in striatum: an implication for sensitivity to heavy ion radiations. Front Cell Neurosci 2024; 18:1356536. [PMID: 38440149 PMCID: PMC10911010 DOI: 10.3389/fncel.2024.1356536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 01/08/2024] [Indexed: 03/06/2024] Open
Abstract
[This corrects the article DOI: 10.3389/fncel.2023.1252958.].
Collapse
Affiliation(s)
- Zixuan Chen
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Medical Technology, Beijing Institute of Technology, Beijing, China
| | - Yumeng Li
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Medical Technology, Beijing Institute of Technology, Beijing, China
| | - Madiha Rasheed
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Medical Technology, Beijing Institute of Technology, Beijing, China
| | - Hao Wang
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Medical Technology, Beijing Institute of Technology, Beijing, China
| | - Runhong Lei
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Medical Technology, Beijing Institute of Technology, Beijing, China
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, China
| | - Tuo Zhao
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Medical Technology, Beijing Institute of Technology, Beijing, China
| | - Yulin Deng
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Medical Technology, Beijing Institute of Technology, Beijing, China
| | - Hong Ma
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Medical Technology, Beijing Institute of Technology, Beijing, China
| |
Collapse
|
28
|
Li Z, Wu J, Zhao T, Wei Y, Xu Y, Liu Z, Li X, Chen X. Microglial activation in spaceflight and microgravity: potential risk of cognitive dysfunction and poor neural health. Front Cell Neurosci 2024; 18:1296205. [PMID: 38425432 PMCID: PMC10902453 DOI: 10.3389/fncel.2024.1296205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 01/29/2024] [Indexed: 03/02/2024] Open
Abstract
Due to the increased crewed spaceflights in recent years, it is vital to understand how the space environment affects human health. A lack of gravitational force is known to risk multiple physiological functions of astronauts, particularly damage to the central nervous system (CNS). As innate immune cells of the CNS, microglia can transition from a quiescent state to a pathological state, releasing pro-inflammatory cytokines that contribute to neuroinflammation. There are reports indicating that microglia can be activated by simulating microgravity or exposure to galactic cosmic rays (GCR). Consequently, microglia may play a role in the development of neuroinflammation during spaceflight. Prolonged spaceflight sessions raise concerns about the chronic activation of microglia, which could give rise to various neurological disorders, posing concealed risks to the neural health of astronauts. This review summarizes the risks associated with neural health owing to microglial activation and explores the stressors that trigger microglial activation in the space environment. These stressors include GCR, microgravity, and exposure to isolation and stress. Of particular focus is the activation of microglia under microgravity conditions, along with the proposal of a potential mechanism.
Collapse
Affiliation(s)
- Zihan Li
- Beijing International Science and Technology Cooperation Base for Antiviral Drugs, College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| | - Jiarui Wu
- Beijing International Science and Technology Cooperation Base for Antiviral Drugs, College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| | - Tianyuan Zhao
- Beijing International Science and Technology Cooperation Base for Antiviral Drugs, College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| | - Yiyun Wei
- Beijing International Science and Technology Cooperation Base for Antiviral Drugs, College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| | - Yajing Xu
- Beijing International Science and Technology Cooperation Base for Antiviral Drugs, College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| | - Zongjian Liu
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Xiaoqiong Li
- School of Life Sciences, Beijing Institute of Technology, Beijing, China
| | - Xuechai Chen
- Beijing International Science and Technology Cooperation Base for Antiviral Drugs, College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| |
Collapse
|
29
|
Smith L. Space station and spacecraft environmental conditions and human mental health: Specific recommendations and guidelines. LIFE SCIENCES IN SPACE RESEARCH 2024; 40:126-134. [PMID: 38245337 DOI: 10.1016/j.lssr.2023.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/18/2023] [Accepted: 10/08/2023] [Indexed: 01/22/2024]
Abstract
The way that a given environment may influence human mental health is widely established, with decades of research linking anxiety, depression, stress, productivity, and general mood with all facets of a given environment, including noise levels, lighting, air quality, and other factors. The environmental conditions of a space habitat have far reaching consequences for human mental health and should be carefully managed. This manuscript serves to briefly review what is known about the main components of a space habitat (e.g., noise levels, lighting, air quality, privacy, plant life, etc.), and provide specific and clear recommendations for mission planners and space habitat designers. Where appropriate, opportunities for future research are highlighted.
Collapse
Affiliation(s)
- Logan Smith
- Oklahoma State University, 116 Psychology Building, Stillwater, OK 74078, USA.
| |
Collapse
|
30
|
Vozenin MC, Alaghband Y, Drayson OGG, Piaget F, Leavitt R, Allen BD, Doan NL, Rostomyan T, Stabilini A, Reggiani D, Hajdas W, Yukihara EG, Norbury JW, Bailat C, Desorgher L, Baulch JE, Limoli CL. More May Not be Better: Enhanced Spacecraft Shielding May Exacerbate Cognitive Decrements by Increasing Pion Exposures during Deep Space Exploration. Radiat Res 2024; 201:93-103. [PMID: 38171489 DOI: 10.1667/rade-23-00241.1.s1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 12/15/2023] [Indexed: 01/05/2024]
Abstract
The pervasiveness of deep space radiation remains a confounding factor for the transit of humans through our solar system. Spacecraft shielding both protects astronauts but also contributes to absorbed dose through galactic cosmic ray interactions that produce secondary particles. The resultant biological effects drop to a minimum for aluminum shielding around 20 g/cm2 but increase with additional shielding. The present work evaluates for the first time, the impact of secondary pions on central nervous system functionality. The fractional pion dose emanating from thicker shielded spacecraft regions could contribute up to 10% of the total absorbed radiation dose. New results from the Paul Scherrer Institute have revealed that low dose exposures to 150 MeV positive and negative pions, akin to a Mars mission, result in significant, long-lasting cognitive impairments. These surprising findings emphasize the need to carefully evaluate shielding configurations to optimize safe exposure limits for astronauts during deep space travel.
Collapse
Affiliation(s)
- Marie-Catherine Vozenin
- Laboratory of Radiation Oncology, Department of Radiation Oncology, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Yasaman Alaghband
- Department of Radiation Oncology, University of California, Irvine, California 92697-2695
| | - Olivia G G Drayson
- Department of Radiation Oncology, University of California, Irvine, California 92697-2695
| | - Filippo Piaget
- Laboratory of Radiation Oncology, Department of Radiation Oncology, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Ron Leavitt
- Laboratory of Radiation Oncology, Department of Radiation Oncology, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Barrett D Allen
- Department of Radiation Oncology, University of California, Irvine, California 92697-2695
| | - Ngoc-Lien Doan
- Department of Radiation Oncology, University of California, Irvine, California 92697-2695
| | | | | | | | | | | | | | - Claude Bailat
- Institute of Radiation Physics, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Laurent Desorgher
- Institute of Radiation Physics, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Janet E Baulch
- Department of Radiation Oncology, University of California, Irvine, California 92697-2695
| | - Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine, California 92697-2695
| |
Collapse
|
31
|
Chen Z, Li Y, Rasheed M, Wang H, Lei R, Zhao T, Deng Y, Ma H. Altered expression of inflammation-associated molecules in striatum: an implication for sensitivity to heavy ion radiations. Front Cell Neurosci 2023; 17:1252958. [PMID: 38107411 PMCID: PMC10725200 DOI: 10.3389/fncel.2023.1252958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 11/06/2023] [Indexed: 12/19/2023] Open
Abstract
Background and objective Heavy ion radiation is one of the major hazards astronauts face during space expeditions, adversely affecting the central nervous system. Radiation causes severe damage to sensitive brain regions, especially the striatum, resulting in cognitive impairment and other physiological issues in astronauts. However, the intensity of brain damage and associated underlying molecular pathological mechanisms mediated by heavy ion radiation are still unknown. The present study is aimed to identify the damaging effect of heavy ion radiation on the striatum and associated underlying pathological mechanisms. Materials and methods Two parallel cohorts of rats were exposed to radiation in multiple doses and times. Cohort I was exposed to 15 Gy of 12C6+ ions radiation, whereas cohort II was exposed to 3.4 Gy and 8 Gy with 56Fe26+ ions irradiation. Physiological and behavioural tests were performed, followed by 18F-FDG-PET scans, transcriptomics analysis of the striatum, and in-vitro studies to verify the interconnection between immune cells and neurons. Results Both cohorts revealed more persistent striatum dysfunction than other brain regions under heavy ion radiation at multiple doses and time, exposed by physiological, behavioural, and 18F-FDG-PET scans. Transcriptomic analysis revealed that striatum dysfunction is linked with an abnormal immune system. In vitro studies demonstrated that radiation mediated diversified effects on different immune cells and sustained monocyte viability but inhibited its differentiation and migration, leading to chronic neuroinflammation in the striatum and might affect other associated brain regions. Conclusion Our findings suggest that striatum dysfunction under heavy ion radiation activates abnormal immune systems, leading to chronic neuroinflammation and neuronal injury.
Collapse
Affiliation(s)
- Zixuan Chen
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Medical Technology, Beijing Institute of Technology, Beijing, China
| | - Yumeng Li
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Medical Technology, Beijing Institute of Technology, Beijing, China
| | - Madiha Rasheed
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Medical Technology, Beijing Institute of Technology, Beijing, China
| | - Hao Wang
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Medical Technology, Beijing Institute of Technology, Beijing, China
| | - Runhong Lei
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Medical Technology, Beijing Institute of Technology, Beijing, China
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, China
| | - Tuo Zhao
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Medical Technology, Beijing Institute of Technology, Beijing, China
| | - Yulin Deng
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Medical Technology, Beijing Institute of Technology, Beijing, China
| | - Hong Ma
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Medical Technology, Beijing Institute of Technology, Beijing, China
| |
Collapse
|
32
|
Desai RI, Kangas BD, Luc OT, Solakidou E, Smith EC, Dawes MH, Ma X, Makriyannis A, Chatterjee S, Dayeh MA, Muñoz-Jaramillo A, Desai MI, Limoli CL. Complex 33-beam simulated galactic cosmic radiation exposure impacts cognitive function and prefrontal cortex neurotransmitter networks in male mice. Nat Commun 2023; 14:7779. [PMID: 38012180 PMCID: PMC10682413 DOI: 10.1038/s41467-023-42173-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 09/28/2023] [Indexed: 11/29/2023] Open
Abstract
Astronauts will encounter extended exposure to galactic cosmic radiation (GCR) during deep space exploration, which could impair brain function. Here, we report that in male mice, acute or chronic GCR exposure did not modify reward sensitivity but did adversely affect attentional processes and increased reaction times. Potassium (K+)-stimulation in the prefrontal cortex (PFC) elevated dopamine (DA) but abolished temporal DA responsiveness after acute and chronic GCR exposure. Unlike acute GCR, chronic GCR increased levels of all other neurotransmitters, with differences evident between groups after higher K+-stimulation. Correlational and machine learning analysis showed that acute and chronic GCR exposure differentially reorganized the connection strength and causation of DA and other PFC neurotransmitter networks compared to controls which may explain space radiation-induced neurocognitive deficits.
Collapse
Affiliation(s)
- Rajeev I Desai
- Department of Psychiatry, Harvard Medical School, Boston, MA, 02115, USA.
- Behavioral Biology Program, McLean Hospital, Belmont, MA, 02478, USA.
- Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, 02115, USA.
| | - Brian D Kangas
- Department of Psychiatry, Harvard Medical School, Boston, MA, 02115, USA
- Behavioral Biology Program, McLean Hospital, Belmont, MA, 02478, USA
| | - Oanh T Luc
- Department of Psychiatry, Harvard Medical School, Boston, MA, 02115, USA
- Behavioral Biology Program, McLean Hospital, Belmont, MA, 02478, USA
| | - Eleana Solakidou
- Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, 02115, USA
- Medical School, University of Crete, Heraklion, Greece
| | - Evan C Smith
- Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, 02115, USA
| | - Monica H Dawes
- Department of Psychiatry, Harvard Medical School, Boston, MA, 02115, USA
- Behavioral Biology Program, McLean Hospital, Belmont, MA, 02478, USA
| | - Xiaoyu Ma
- Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, 02115, USA
| | - Alexandros Makriyannis
- Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, 02115, USA
| | | | - Maher A Dayeh
- Southwest Research Institute, San Antonio, TX, 78238, USA
- University of San Antonio, San Antonio, TX, 78249, USA
| | | | - Mihir I Desai
- Southwest Research Institute, San Antonio, TX, 78238, USA
- University of San Antonio, San Antonio, TX, 78249, USA
| | - Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine, Orange, CA, 92697, USA
| |
Collapse
|
33
|
Pan C, Zhang Y, Yan J, Zhou Y, Wang S, Liu X, Zhang P, Yang H. Extreme environments and human health: From the immune microenvironments to immune cells. ENVIRONMENTAL RESEARCH 2023; 236:116800. [PMID: 37527745 DOI: 10.1016/j.envres.2023.116800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 07/20/2023] [Accepted: 07/29/2023] [Indexed: 08/03/2023]
Abstract
Exposure to extreme environments causes specific acute and chronic physiological responses in humans. The adaptation and the physiological processes under extreme environments predominantly affect multiple functional systems of the organism, in particular, the immune system. Dysfunction of the immune system affected by several extreme environments (including hyperbaric environment, hypoxia, blast shock, microgravity, hypergravity, radiation exposure, and magnetic environment) has been observed from clinical macroscopic symptoms to intracorporal immune microenvironments. Therefore, simulated extreme conditions are engineered for verifying the main influenced characteristics and factors in the immune microenvironments. This review summarizes the responses of immune microenvironments to these extreme environments during in vivo or in vitro exposure, and the approaches of engineering simulated extreme environments in recent decades. The related microenvironment engineering, signaling pathways, molecular mechanisms, clinical therapy, and prevention strategies are also discussed.
Collapse
Affiliation(s)
- Chengwei Pan
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, China; Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Yuzhi Zhang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, China; Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Jinxiao Yan
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, China; Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Yidan Zhou
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, China; Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Sijie Wang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, China; Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Xiru Liu
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, China; Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Pan Zhang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; School of Food Science and Engineering, Shaanxi University of Science & Technology, 710021, China.
| | - Hui Yang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, China; Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China.
| |
Collapse
|
34
|
Desorgher L, Stabilini A, Rostomyan T, Reggiani D, Hajdas W, Marcinkowski RM, Vozenin MC, Limoli CL, Yukihara EG, Bailat C. Dosimetry of the PIM1 Pion Beam at the Paul Scherrer Institute for Radiobiological Studies of Mice. Radiat Res 2023; 200:357-365. [PMID: 37702413 DOI: 10.1667/rade-23-00029.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 07/31/2023] [Indexed: 09/14/2023]
Abstract
Significant past work has identified unexpected risks of central nervous system (CNS) exposure to the space radiation environment, where long-lasting functional decrements have been associated with multiple ion species delivered at low doses and dose rates. As shielding is the only established intervention capable of limiting exposure to the dangerous radiation fields in space, the recent discovery that pions, emanating from regions of enhanced shielding, can contribute significantly to the total absorbed dose on a deep space mission poses additional concerns. As a prerequisite to biological studies evaluating pion dose equivalents for various CNS exposure scenarios of mice, a careful dosimetric validation study is required. Within our ultimate goal of evaluating the functional consequences of defined pion exposures to CNS functionality, we report in this article the detailed dosimetry of the PiMI pion beam line at the Paul Scherrer Institute, which was developed in support of radiobiological experiments. Beam profiles and contamination of the beam by protons, electrons, positrons and muons were characterized prior to the mice irradiations. The dose to the back and top of the mice was measured using thermoluminescent dosimeters (TLD) and optically simulated luminescence (OSL) to cross-validate the dosimetry results. Geant4 Monte Carlo simulations of radiation exposure of a mouse phantom in water by charged pions were also performed to quantify the difference between the absorbed dose from the OSL and TLD and the absorbed dose to water, using a simple model of the mouse brain. The absorbed dose measured by the OSL dosimeters and TLDs agreed within 5-10%. A 30% difference between the measured absorbed dose and the dose calculated by Geant4 in the dosimeters was obtained, probably due to the approximated Monte Carlo configuration compared to the experiment. A difference of 15-20% between the calculated absorbed dose to water at a 5 mm depth and in the passive dosimeters was obtained, suggesting the need for a correction factor of the measured dose to obtain the absorbed dose in the mouse brain. Finally, based on the comparison of the experimental data and the Monte Carlo calculations, we consider the dose measurement to be accurate to within 15-20%.
Collapse
Affiliation(s)
- L Desorgher
- Institute of Radiation Physics, Lausanne University Hospital and Lausanne University, Switzerland
| | - A Stabilini
- Paul Scherrer Institute (PSI), Villigen, Switzerland
| | - T Rostomyan
- Paul Scherrer Institute (PSI), Villigen, Switzerland
| | - D Reggiani
- Paul Scherrer Institute (PSI), Villigen, Switzerland
| | - W Hajdas
- Paul Scherrer Institute (PSI), Villigen, Switzerland
| | - R M Marcinkowski
- Paul Scherrer Institute (PSI), Villigen, Switzerland
- SE2S GMBH, Boppelsen ZH, Switzerland
| | - M-C Vozenin
- CHUV-Radiation-oncology Laboratory, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - C L Limoli
- Department of Radiation Oncology, University of California, Irvine, California
| | - E G Yukihara
- Paul Scherrer Institute (PSI), Villigen, Switzerland
| | - C Bailat
- Institute of Radiation Physics, Lausanne University Hospital and Lausanne University, Switzerland
| |
Collapse
|
35
|
Britten RA, Fesshaye A, Tidmore A, Liu A, Blackwell AA. Loss of Cognitive Flexibility Practice Effects in Female Rats Exposed to Simulated Space Radiation. Radiat Res 2023; 200:256-265. [PMID: 37527363 DOI: 10.1667/rade-22-00196.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 06/27/2023] [Indexed: 08/03/2023]
Abstract
During the planned missions to Mars, astronauts will be faced with many potential health hazards including prolonged exposure to space radiation. Ground-based studies have shown that exposure to space radiation impairs the performance of male rats in cognitive flexibility tasks which involve processes that are essential to rapidly and efficiently adapting to different situations. However, there is presently a paucity of information on the effects of space radiation on cognitive flexibility in female rodents. This study has determined the impact that exposure to a low (10 cGy) dose of ions from the simplified 5-ion galactic cosmic ray simulation [https://www.bnl.gov/nsrl/userguide/SimGCRSim.php (07/2023)] (GCRSim) beam or 250 MeV/n 4He ions has on the ability of female Wistar rats to perform in constrained [attentional set shifting (ATSET)] and unconstrained cognitive flexibility (UCFlex) tasks. Female rats exposed to GCRSim exhibited multiple decrements in ATSET performance. Firstly, GCRSim exposure impaired performance in the compound discrimination (CD) stage of the ATSET task. While the ability of rats to identify the rewarded cue was not compromised, the time the rats required to do so significantly increased. Secondly, both 4He and GCRSim exposure reduced the ability of rats to reach criterion in the compound discrimination reversal (CDR) stage. Approximately 20% of the irradiated rats were unable to complete the CDR task; furthermore, the irradiated rats that did reach criterion took more attempts to do so than did the sham-treated animals. Radiation exposure also altered the magnitude and/or nature of practice effects. A comparison of performance metrics from the pre-screen and post-exposure ATSET task revealed that while the sham-treated rats completed the post-exposure CD stage of the ATSET task in 30% less time than for completion of the pre-screen ATSET task, the irradiated rats took 30-50% longer to do so. Similarly, while sham-treated rats completed the CDR stage in ∼10% fewer attempts in the post-exposure task compared to the pre-screen task, in contrast, the 4He- and GCRSim-exposed cohorts took more (∼2-fold) attempts to reach criterion in the post-exposure task than in the pre-screen task. In conclusion, this study demonstrates that female rats are susceptible to radiation-induced loss of performance in the constrained ATSET cognitive flexibility task. Moreover, exposure to radiation leads to multiple performance decrements, including loss of practice effects, an increase in anterograde interference and reduced ability or unwillingness to switch attention. Should similar effects occur in humans, astronauts may have a compromised ability to perform complex tasks.
Collapse
Affiliation(s)
- Richard A Britten
- EVMS Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507
- EVMS Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, Virginia 23507
- Center for Integrative Neuroscience and Inflammatory diseases, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Arriyam Fesshaye
- EVMS Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Alyssa Tidmore
- EVMS Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Aiyi Liu
- EVMS Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Ashley A Blackwell
- EVMS Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507
- Center for Integrative Neuroscience and Inflammatory diseases, Eastern Virginia Medical School, Norfolk, Virginia 23507
| |
Collapse
|
36
|
Babu B, Pawar S, Mittal A, Kolanthai E, Neal CJ, Coathup M, Seal S. Nanotechnology enabled radioprotectants to reduce space radiation-induced reactive oxidative species. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1896. [PMID: 37190884 DOI: 10.1002/wnan.1896] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 04/04/2023] [Accepted: 04/18/2023] [Indexed: 05/17/2023]
Abstract
Interest in space exploration has seen substantial growth following recent launch and operation of modern space technologies. In particular, the possibility of travel beyond low earth orbit is seeing sustained support. However, future deep space travel requires addressing health concerns for crews under continuous, longer-term exposure to adverse environmental conditions. Among these challenges, radiation-induced health issues are a major concern. Their potential to induce chronic illness is further potentiated by the microgravity environment. While investigations into the physiological effects of space radiation are still under investigation, studies on model ionizing radiation conditions, in earth and micro-gravity conditions, can provide needed insight into relevant processes. Substantial formation of high, sustained reactive oxygen species (ROS) evolution during radiation exposure is a clear threat to physiological health of space travelers, producing indirect damage to various cell structures and requiring therapeutic address. Radioprotection toward the skeletal system components is essential to astronaut health, due to the high radio-absorption cross-section of bone mineral and local hematopoiesis. Nanotechnology can potentially function as radioprotectant and radiomitigating agents toward ROS and direct radiation damage. Nanoparticle compositions such as gold, silver, platinum, carbon-based materials, silica, transition metal dichalcogenides, and ceria have all shown potential as viable radioprotectants to mitigate space radiation effects with nanoceria further showing the ability to protect genetic material from oxidative damage in several studies. As research into space radiation-induced health problems develops, this review intends to provide insights into the nanomaterial design to ameliorate pathological effects from ionizing radiation exposure. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Nanotechnology Approaches to Biology > Cells at the Nanoscale Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Balaashwin Babu
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, Florida, USA
- Nanoscience Technology Center, University of Central Florida, Orlando, Florida, USA
| | - Shreya Pawar
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, Florida, USA
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| | - Agastya Mittal
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, Florida, USA
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| | - Elayaraja Kolanthai
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, Florida, USA
| | - Craig J Neal
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, Florida, USA
| | - Melanie Coathup
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, Florida, USA
| | - Sudipta Seal
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, Florida, USA
- College of Medicine, Nanoscience Technology Center, University of Central Florida, Orlando, Florida, USA
| |
Collapse
|
37
|
Impey S, Pelz C, Riparip LK, Tafessu A, Fareh F, Zuloaga DG, Marzulla T, Stewart B, Rosi S, Turker MS, Raber J. Postsynaptic density radiation signature following space irradiation. Front Physiol 2023; 14:1215535. [PMID: 37440997 PMCID: PMC10334289 DOI: 10.3389/fphys.2023.1215535] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 06/14/2023] [Indexed: 07/15/2023] Open
Abstract
Introduction: The response of the brain to space radiation is an important concern for astronauts during space missions. Therefore, we assessed the response of the brain to 28Si ion irradiation (600 MeV/n), a heavy ion present in the space environment, on cognitive performance and whether the response is associated with altered DNA methylation in the hippocampus, a brain area important for cognitive performance. Methods: We determined the effects of 28Si ion irradiation on object recognition, 6-month-old mice irradiated with 28Si ions (600 MeV/n, 0.3, 0.6, and 0.9 Gy) and cognitively tested two weeks later. In addition, we determined if those effects were associated with alterations in hippocampal networks and/or hippocampal DNA methylation. Results: At 0.3 Gy, but not at 0.6 Gy or 0.9 Gy, 28Si ion irradiation impaired cognition that correlated with altered gene expression and 5 hmC profiles that mapped to specific gene ontology pathways. Comparing hippocampal DNA hydroxymethylation following proton, 56Fe ion, and 28Si ion irradiation revealed a general space radiation synaptic signature with 45 genes that are associated with profound phenotypes. The most significant categories were glutamatergic synapse and postsynaptic density. Discussion: The brain's response to space irradiation involves novel excitatory synapse and postsynaptic remodeling.
Collapse
Affiliation(s)
- Soren Impey
- Department of Pediatrics, Oregon Stem Cell Center, Oregon Health and Science University, Portland, OR, United States
- Dow Neuroscience Laboratories, Department of Cell and Developmental Biology, Legacy Research Institute, Legacy Health Systems, Oregon Health and Science University, Portland, OR, United States
| | - Carl Pelz
- Department of Pediatrics, Oregon Stem Cell Center, Oregon Health and Science University, Portland, OR, United States
| | - Lara-Kirstie Riparip
- Departments of Neurological Surgery and Physical Therapy and Rehabilitation Science, Brain and Spinal Injury Center, University of California, San Francisco, San Francisco, CA, United States
| | - Amanuel Tafessu
- Department of Pediatrics, Oregon Stem Cell Center, Oregon Health and Science University, Portland, OR, United States
| | - Fatema Fareh
- Department of Pediatrics, Oregon Stem Cell Center, Oregon Health and Science University, Portland, OR, United States
| | - Damian G. Zuloaga
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, United States
| | - Tessa Marzulla
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, United States
| | - Blair Stewart
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, United States
| | - Susanna Rosi
- Departments of Neurological Surgery and Physical Therapy and Rehabilitation Science, Brain and Spinal Injury Center, University of California, San Francisco, San Francisco, CA, United States
| | - Mitchell S. Turker
- Department of Molecular and Medical Genetics, Oregon Institute of Occupational Health Sciences, Oregon Health and Science University, Portland, OR, United States
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, United States
- Departments of Neurology and Radiation Medicine, Division of Neuroscience ONPRC, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
38
|
Britten RA, Limoli CL. New Radiobiological Principles for the CNS Arising from Space Radiation Research. Life (Basel) 2023; 13:1293. [PMID: 37374076 DOI: 10.3390/life13061293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/17/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
Traditionally, the brain has been regarded as a relatively insensitive late-reacting tissue, with radiologically detectable damage not being reported at doses < 60 Gy. When NASA proposed interplanetary exploration missions, it was required to conduct an intensive health and safety evaluation of cancer, cardiovascular, and cognitive risks associated with exposure to deep space radiation (SR). The SR dose that astronauts on a mission to Mars are predicted to receive is ~300 mGy. Even after correcting for the higher RBE of the SR particles, the biologically effective SR dose (<1 Gy) would still be 60-fold lower than the threshold dose for clinically detectable neurological damage. Unexpectedly, the NASA-funded research program has consistently reported that low (<250 mGy) doses of SR induce deficits in multiple cognitive functions. This review will discuss these findings and the radical paradigm shifts in radiobiological principles for the brain that were required in light of these findings. These included a shift from cell killing to loss of function models, an expansion of the critical brain regions for radiation-induced cognitive impediments, and the concept that the neuron may not be the sole critical target for neurocognitive impairment. The accrued information on how SR exposure impacts neurocognitive performance may provide new opportunities to reduce neurocognitive impairment in brain cancer patients.
Collapse
Affiliation(s)
- Richard A Britten
- EVMS Radiation Oncology, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Charles L Limoli
- Department Radiation Oncology, University of California-Irvine, Irvine, CA 92697, USA
| |
Collapse
|
39
|
Puukila S, Siu O, Rubinstein L, Tahimic CGT, Lowe M, Tabares Ruiz S, Korostenskij I, Semel M, Iyer J, Mhatre SD, Shirazi-Fard Y, Alwood JS, Paul AM, Ronca AE. Galactic Cosmic Irradiation Alters Acute and Delayed Species-Typical Behavior in Male and Female Mice. Life (Basel) 2023; 13:life13051214. [PMID: 37240858 DOI: 10.3390/life13051214] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/14/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023] Open
Abstract
Exposure to space galactic cosmic radiation is a principal consideration for deep space missions. While the effects of space irradiation on the nervous system are not fully known, studies in animal models have shown that exposure to ionizing radiation can cause neuronal damage and lead to downstream cognitive and behavioral deficits. Cognitive health implications put humans and missions at risk, and with the upcoming Artemis missions in which female crew will play a major role, advance critical analysis of the neurological and performance responses of male and female rodents to space radiation is vital. Here, we tested the hypothesis that simulated Galactic Cosmic Radiation (GCRSim) exposure disrupts species-typical behavior in mice, including burrowing, rearing, grooming, and nest-building that depend upon hippocampal and medial prefrontal cortex circuitry. Behavior comprises a remarkably well-integrated representation of the biology of the whole animal that informs overall neural and physiological status, revealing functional impairment. We conducted a systematic dose-response analysis of mature (6-month-old) male and female mice exposed to either 5, 15, or 50 cGy 5-ion GCRSim (H, Si, He, O, Fe) at the NASA Space Radiation Laboratory (NSRL). Behavioral performance was evaluated at 72 h (acute) and 91-days (delayed) postradiation exposure. Specifically, species-typical behavior patterns comprising burrowing, rearing, and grooming as well as nest building were analyzed. A Neuroscore test battery (spontaneous activity, proprioception, vibrissae touch, limb symmetry, lateral turning, forelimb outstretching, and climbing) was performed at the acute timepoint to investigate early sensorimotor deficits postirradiation exposure. Nest construction, a measure of neurological and organizational function in rodents, was evaluated using a five-stage Likert scale 'Deacon' score that ranged from 1 (a low score where the Nestlet is untouched) to 5 (a high score where the Nestlet is completely shredded and shaped into a nest). Differential acute responses were observed in females relative to males with respect to species-typical behavior following 15 cGy exposure while delayed responses were observed in female grooming following 50 cGy exposure. Significant sex differences were observed at both timepoints in nest building. No deficits in sensorimotor behavior were observed via the Neuroscore. This study revealed subtle, sexually dimorphic GCRSim exposure effects on mouse behavior. Our analysis provides a clearer understanding of GCR dose effects on species typical, sensorimotor and organizational behaviors at acute and delayed timeframes postirradiation, thereby setting the stage for the identification of underlying cellular and molecular events.
Collapse
Affiliation(s)
- Stephanie Puukila
- Oak Ridge Associated Universities, Oak Ridge, TN 37831, USA
- NASA, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Olivia Siu
- Space Life Sciences Training Program (SLSTP), NASA Ames Research Center, Moffett Field, CA 94035, USA
- Department of Human Factors and Behavioral Neurobiology, Embry-Riddle Aeronautical University, Daytona Beach, FL 32114, USA
| | - Linda Rubinstein
- Universities Space Research Association, Columbia, MD 21046, USA
- The Joseph Sagol Neuroscience Center, Sheba Hospital, Ramat Gan 52621, Israel
| | - Candice G T Tahimic
- NASA, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
- Department of Biology, University of North Florida, Jacksonville, FL 32224, USA
| | - Moniece Lowe
- NASA, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
- Blue Marble Space Institute of Science, Seattle, WA 98154, USA
| | - Steffy Tabares Ruiz
- NASA, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
- Blue Marble Space Institute of Science, Seattle, WA 98154, USA
| | - Ivan Korostenskij
- Department of Biology, University of North Florida, Jacksonville, FL 32224, USA
| | - Maya Semel
- Department of Biology, University of North Florida, Jacksonville, FL 32224, USA
| | - Janani Iyer
- NASA, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
- Universities Space Research Association, Columbia, MD 21046, USA
- KBR, Houston, TX 77002, USA
| | - Siddhita D Mhatre
- NASA, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
- KBR, Houston, TX 77002, USA
| | - Yasaman Shirazi-Fard
- NASA, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Joshua S Alwood
- NASA, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Amber M Paul
- NASA, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
- Department of Human Factors and Behavioral Neurobiology, Embry-Riddle Aeronautical University, Daytona Beach, FL 32114, USA
- Blue Marble Space Institute of Science, Seattle, WA 98154, USA
| | - April E Ronca
- NASA, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
- Wake Forest Medical School, Winston-Salem, NC 27101, USA
| |
Collapse
|
40
|
Ruprecht NA, Singhal S, Schaefer K, Gill JS, Bansal B, Sens D, Singhal SK. Establishing a genomic radiation-age association for space exploration supplements lung disease differentiation. Front Public Health 2023; 11:1161124. [PMID: 37250098 PMCID: PMC10213902 DOI: 10.3389/fpubh.2023.1161124] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 04/07/2023] [Indexed: 05/31/2023] Open
Abstract
Purpose One possible way to quantify each individual's response or damage from ionizing radiation is to estimate their accelerated biological age following exposure. Since there is currently no definitive way to know if biological age estimations are accurate, we aim to establish a rad-age association using genomics as its foundation. Methods Two datasets were combined and used to empirically find the age cutoff between young and old patients. With age as both a categorical and continuous variable, two other datasets that included radiation exposure are used to test the interaction between radiation and age. The gene lists are oriented in preranked lists for both pathway and diseases analysis. Finally, these genes are used to evaluate another dataset on the clinical relevance in differentiating lung disease given ethnicity and sex using both pairwise t-tests and linear models. Results Using 12 well-known genes associated with aging, a threshold of 29-years-old was found to be the difference between young and old patients. The two interaction tests yielded 234 unique genes such that pathway analysis flagged IL-1 signaling and PRPP biosynthesis as significant with high cell proliferation diseases and carcinomas being a common trend. LAPTM4B was the only gene with significant interaction among lung disease, ethnicity, and sex, with fold change greater than two. Conclusion The results corroborate an initial association between radiation and age, given inflammation and metabolic pathways and multiple genes emphasizing mitochondrial function, oxidation, and histone modification. Being able to tie rad-age genes to lung disease supplements future work for risk assessment following radiation exposure.
Collapse
Affiliation(s)
- Nathan A. Ruprecht
- Department of Biomedical Engineering, University of North Dakota, Grand Forks, ND, United States
| | - Sonalika Singhal
- Department of Pathology, University of North Dakota, Grand Forks, ND, United States
| | - Kalli Schaefer
- Department of Biomedical Engineering, University of North Dakota, Grand Forks, ND, United States
| | - Jappreet S. Gill
- Department of Biomedical Engineering, University of North Dakota, Grand Forks, ND, United States
| | - Benu Bansal
- Department of Biomedical Engineering, University of North Dakota, Grand Forks, ND, United States
| | - Donald Sens
- Department of Pathology, University of North Dakota, Grand Forks, ND, United States
| | - Sandeep K. Singhal
- Department of Biomedical Engineering, University of North Dakota, Grand Forks, ND, United States
- Department of Pathology, University of North Dakota, Grand Forks, ND, United States
| |
Collapse
|
41
|
Stephenson S, Liu A, Blackwell AA, Britten RA. Multiple decrements in switch task performance in female rats exposed to space radiation. Behav Brain Res 2023; 449:114465. [PMID: 37142163 DOI: 10.1016/j.bbr.2023.114465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/14/2023] [Accepted: 05/01/2023] [Indexed: 05/06/2023]
Abstract
Astronauts on the Artemis missions to the Moon and Mars will be exposed to unavoidable Galactic Cosmic Radiation (GCR). Studies using male rats suggest that GCR exposure impairs several processes required for cognitive flexibility performance, including attention and task switching. Currently no comparable studies have been conducted with female rats. Given that both males and females will travel into deep space, this study determined whether simulated GCR (GCRsim) exposure impairs task switching performance in female rats. Female Wistar rats exposed to 10cGy GCRsim (n = 12) and shams (n=14) were trained to perform a touchscreen-based switch task that mimics a switch task used to evaluate pilots' response times. In comparison to sham rats, three-fold more GCRsim-exposed rats failed to complete the stimulus response stage of training, a high cognitive loading task. In the switch task, 50% of the GCRsim-exposed rats failed to consistently transition between the repeated and switch blocks of stimuli, which they completed during lower cognitive loading training stages. The GCRsim-exposed rats that completed the switch task only performed at 65% of the accuracy of shams. Female rats exposed to GCRsim thus exhibit multiple decrements in the switch task under high, but not low, cognitive loading conditions. While the operational significance of this performance decrement is unknown, if GCRSim exposure was to induce similar effects in astronauts, our data suggests there may be a reduced ability to execute task switching under high cognitive loading situations.
Collapse
Affiliation(s)
- Samuel Stephenson
- School of Medicine, Eastern Virginia Medical School, Norfolk, Virginia 23507 USA
| | - Aiyi Liu
- EVMS Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507 USA
| | - Ashley A Blackwell
- EVMS Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507 USA; Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, Virginia 23507 USA
| | - Richard A Britten
- EVMS Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507 USA; Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, Virginia 23507 USA.
| |
Collapse
|
42
|
Roggan MD, Kronenberg J, Wollert E, Hoffmann S, Nisar H, Konda B, Diegeler S, Liemersdorf C, Hellweg CE. Unraveling astrocyte behavior in the space brain: Radiation response of primary astrocytes. Front Public Health 2023; 11:1063250. [PMID: 37089489 PMCID: PMC10116417 DOI: 10.3389/fpubh.2023.1063250] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 03/06/2023] [Indexed: 04/09/2023] Open
Abstract
IntroductionExposure to space conditions during crewed long-term exploration missions can cause several health risks for astronauts. Space radiation, isolation and microgravity are major limiting factors. The role of astrocytes in cognitive disturbances by space radiation is unknown. Astrocytes' response toward low linear energy transfer (LET) X-rays and high-LET carbon (12C) and iron (56Fe) ions was compared to reveal possible effects of space-relevant high-LET radiation. Since astronauts are exposed to ionizing radiation and microgravity during space missions, the effect of simulated microgravity on DNA damage induction and repair was investigated.MethodsPrimary murine cortical astrocytes were irradiated with different doses of X-rays, 12C and 56Fe ions at the heavy ion accelerator GSI. DNA damage and repair (γH2AX, 53BP1), cell proliferation (Ki-67), astrocytes' reactivity (GFAP) and NF-κB pathway activation (p65) were analyzed by immunofluorescence microscopy. Cell cycle progression was investigated by flow cytometry of DNA content. Gene expression changes after exposure to X- rays were investigated by mRNA-sequencing. RT-qPCR for several genes of interest was performed with RNA from X-rays- and heavy-ion-irradiated astrocytes: Cdkn1a, Cdkn2a, Gfap, Tnf, Il1β, Il6, and Tgfβ1. Levels of the pro inflammatory cytokine IL-6 were determined using ELISA. DNA damage response was investigated after exposure to X-rays followed by incubation on a 2D clinostat to simulate the conditions of microgravity.ResultsAstrocytes showed distinct responses toward the three different radiation qualities. Induction of radiation-induced DNA double strand breaks (DSBs) and the respective repair was dose-, LET- and time-dependent. Simulated microgravity had no significant influence on DNA DSB repair. Proliferation and cell cycle progression was not affected by radiation qualities examined in this study. Astrocytes expressed IL-6 and GFAP with constitutive NF-κB activity independent of radiation exposure. mRNA sequencing of X-irradiated astrocytes revealed downregulation of 66 genes involved in DNA damage response and repair, mitosis, proliferation and cell cycle regulation.DiscussionIn conclusion, primary murine astrocytes are DNA repair proficient irrespective of radiation quality. Only minor gene expression changes were observed after X-ray exposure and reactivity was not induced. Co-culture of astrocytes with microglial cells, brain organoids or organotypic brain slice culture experiments might reveal whether astrocytes show a more pronounced radiation response in more complex network architectures in the presence of other neuronal cell types.
Collapse
Affiliation(s)
- Marie Denise Roggan
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
| | - Jessica Kronenberg
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
- Microgravity User Support Center (MUSC), German Aerospace Center (DLR), Cologne, Germany
| | - Esther Wollert
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
| | - Sven Hoffmann
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
- Department of Gravitational Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
| | - Hasan Nisar
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
- Department of Medical Sciences, Pakistan Institute of Engineering and Applied Sciences (PIEAS), Islamabad, Pakistan
| | - Bikash Konda
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
| | - Sebastian Diegeler
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, TX, United States
| | - Christian Liemersdorf
- Department of Gravitational Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
| | - Christine E. Hellweg
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
- *Correspondence: Christine E. Hellweg
| |
Collapse
|
43
|
Drayson OGG, Vozenin MC, Limoli CL. A rigorous behavioral testing platform for the assessment of radiation-induced neurological outcomes. Methods Cell Biol 2023; 180:177-197. [PMID: 37890929 PMCID: PMC11093273 DOI: 10.1016/bs.mcb.2023.02.015] [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: 10/29/2023]
Abstract
Behavioral testing is a popular and reliable method of neurocognitive assessment of rodents but the lack of standard operating procedures has led to a high variation of protocols in use. Therefore, there exists a strong need to standardize protocols for a combined behavioral platform in order to maintain consistency across institutions and assist newcomers in the field. This paper provides details on the methodology of several behavioral tasks which have been validated in identifying radiation induced cognitive impairment as well as provide guidance on timescales and best practices. The cognitive assessments outlined here are optimized for rodent studies and either target learning and memory (open field task, object in updated location, novel object recognition, object in place, and temporal order) or mood and cognition (social interaction, elevated plus maze, light dark box, forced swim test, and fear extinction). We have utilized this platform successfully in evaluating cognitive injury induced by various radiation types, doses, fractionation schedules and also with ultra-high dose rate FLASH radiotherapy. Recommended materials and software are provided as well as advice on methods of data analysis. In this way a comprehensive behavioral platform is described with broad applicability to assess cognitive endpoints critical to therapeutic outcome.
Collapse
Affiliation(s)
- Olivia G G Drayson
- Department of Radiation Oncology, University of California at Irvine, Irvine, CA, United States
| | - Marie-Catherine Vozenin
- Department of Radiation Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.
| | - Charles L Limoli
- Department of Radiation Oncology, University of California at Irvine, Irvine, CA, United States
| |
Collapse
|
44
|
Differential Impact of Social Isolation and Space Radiation on Behavior and Motor Learning in Rats. Life (Basel) 2023; 13:life13030826. [PMID: 36983981 PMCID: PMC10057568 DOI: 10.3390/life13030826] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/10/2023] [Accepted: 03/17/2023] [Indexed: 03/22/2023] Open
Abstract
Future missions to Mars will expose astronauts to several physical and psychological challenges, including exposure to space radiation (SR) and periods of social isolation (SI). Each of these stressors, in addition to mission demands, can affect physical and mental health and potentially negatively impact sleep. The effects of inflight stressors may vary with duration and time course, may be additive or compounding, and may vary with individual differences in stress resilience and vulnerability. Determining how individual differences in resilient and vulnerable phenotypes respond to these mission-related stressors and their interactions with sleep will be crucial for understanding and mitigating factors that can impair performance and damage health. Here, we examined the single and compound effects of ground-based analogs of SI and SR on sensorimotor performance on the balance beam (BB) in rats. We also assessed emotional responses during testing on the BB and assessed whether sensorimotor performance and emotion varied with individual differences in stress resiliency using our established animal model in which stress produces different effects on sleep. Results showed differential motor performance and emotion in the BB task between SI and SR, and these varied based on resilient and vulnerable phenotypes. These findings demonstrate that identifying individual responses to stressors that can impact sensorimotor ability and behavior necessary to perform mission-related tasks will be of particular importance for astronauts and future missions. Should similar effects occur in humans, there may be considerable inter-individual variability in the impact that flight stressors have on the mental health of astronauts and their ability to perform mission-related tasks.
Collapse
|
45
|
The Effects of Galactic Cosmic Rays on the Central Nervous System: From Negative to Unexpectedly Positive Effects That Astronauts May Encounter. BIOLOGY 2023; 12:biology12030400. [PMID: 36979092 PMCID: PMC10044754 DOI: 10.3390/biology12030400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/23/2023] [Accepted: 02/25/2023] [Indexed: 03/06/2023]
Abstract
Galactic cosmic rays (GCR) pose a serious threat to astronauts’ health during deep space missions. The possible functional alterations of the central nervous system (CNS) under GCR exposure can be critical for mission success. Despite the obvious negative effects of ionizing radiation, a number of neutral or even positive effects of GCR irradiation on CNS functions were revealed in ground-based experiments with rodents and primates. This review is focused on the GCR exposure effects on emotional state and cognition, emphasizing positive effects and their potential mechanisms. We integrate these data with GCR effects on adult neurogenesis and pathological protein aggregation, forming a complete picture. We conclude that GCR exposure causes multidirectional effects on cognition, which may be associated with emotional state alterations. However, the irradiation in space-related doses either has no effect or has performance enhancing effects in solving high-level cognition tasks and tasks with a high level of motivation. We suppose the model of neurotransmission changes after irradiation, although the molecular mechanisms of this phenomenon are not fully understood.
Collapse
|
46
|
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: 5] [Impact Index Per Article: 2.5] [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
|
47
|
Cekanaviciute E, Tran D, Nguyen H, Lopez Macha A, Pariset E, Langley S, Babbi G, Malkani S, Penninckx S, Schisler JC, Nguyen T, Karpen GH, Costes SV. Mouse genomic associations with in vitro sensitivity to simulated space radiation. LIFE SCIENCES IN SPACE RESEARCH 2023; 36:47-58. [PMID: 36682829 DOI: 10.1016/j.lssr.2022.07.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 07/23/2022] [Accepted: 07/26/2022] [Indexed: 06/17/2023]
Abstract
Exposure to ionizing radiation is considered by NASA to be a major health hazard for deep space exploration missions. Ionizing radiation sensitivity is modulated by both genomic and environmental factors. Understanding their contributions is crucial for designing experiments in model organisms, evaluating the risk of deep space (i.e. high-linear energy transfer, or LET, particle) radiation exposure in astronauts, and also selecting therapeutic irradiation regimes for cancer patients. We identified single nucleotide polymorphisms in 15 strains of mice, including 10 collaborative cross model strains and 5 founder strains, associated with spontaneous and ionizing radiation-induced in vitro DNA damage quantified based on immunofluorescent tumor protein p53 binding protein (53BP1) positive nuclear foci. Statistical analysis suggested an association with pathways primarily related to cellular signaling, metabolism, tumorigenesis and nervous system damage. We observed different genomic associations in early (4 and 8 h) responses to different LET radiation, while later (24 hour) DNA damage responses showed a stronger overlap across all LETs. Furthermore, a subset of pathways was associated with spontaneous DNA damage, suggesting 53BP1 positive foci as a potential biomarker for DNA integrity in mouse models. Our results suggest several mouse strains as new models to further study the impact of ionizing radiation and validate the identified genetic loci. We also highlight the importance of future human in vitro studies to refine the association of genes and pathways with the DNA damage response to ionizing radiation and identify targets for space travel countermeasures.
Collapse
Affiliation(s)
- Egle Cekanaviciute
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Duc Tran
- Department of Computer Science and Engineering, University of Nevada, Reno, NV 89557, USA
| | - Hung Nguyen
- Department of Computer Science and Engineering, University of Nevada, Reno, NV 89557, USA
| | - Alejandra Lopez Macha
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA; Blue Marble Space Institute of Science, 600 1st Avenue, 1st Floor, Seattle, WA 98104, USA
| | - Eloise Pariset
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA; Universities Space Research Association, 615 National Avenue, Mountain View, CA 94043, USA
| | - Sasha Langley
- Molecular and Cell Biology, UC Berkeley, Berkeley, CA 94720, USA, and Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, 1 Cyclotron Rd, Berkeley, CA 94720, USA
| | - Giulia Babbi
- Bologna Biocomputing Group, FABIT, University of Bologna, Via Belmeloro 6, Bologna, Italy
| | - Sherina Malkani
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA; Blue Marble Space Institute of Science, 600 1st Avenue, 1st Floor, Seattle, WA 98104, USA
| | - Sébastien Penninckx
- Molecular and Cell Biology, UC Berkeley, Berkeley, CA 94720, USA, and Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, 1 Cyclotron Rd, Berkeley, CA 94720, USA; Medical Physics Department, Jules Bordet Institute, Université Libre de Bruxelles, 90 Rue Meylemeersch, 1070 Brussels, Belgium
| | - Jonathan C Schisler
- McAllister Heart Institute and Department of Pharmacology, The University of North Carolina at Chapel Hill, NC 27599, USA
| | - Tin Nguyen
- Department of Computer Science and Engineering, University of Nevada, Reno, NV 89557, USA
| | - Gary H Karpen
- Molecular and Cell Biology, UC Berkeley, Berkeley, CA 94720, USA, and Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, 1 Cyclotron Rd, Berkeley, CA 94720, USA
| | - Sylvain V Costes
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA.
| |
Collapse
|
48
|
Gupta U, Baig S, Majid A, Bell SM. The neurology of space flight; How does space flight effect the human nervous system? LIFE SCIENCES IN SPACE RESEARCH 2023; 36:105-115. [PMID: 36682819 DOI: 10.1016/j.lssr.2022.09.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 09/09/2022] [Accepted: 09/12/2022] [Indexed: 06/17/2023]
Abstract
RATIONALE AND HYPOTHESIS Advancements in technology, human adaptability, and funding have increased space exploration and in turn commercial spaceflight. Corporations such as Space X and Blue Origin are exploring methods to make space tourism possible. This could lead to an increase in the number of patients presenting with neurological diseases associated with spaceflight. Therefore, a comprehensive understanding of spaceflight stressors is required to manage neurological disease in high-risk individuals. OBJECTIVES This review aims to describe the neurological effects of spaceflight and to assess countermeasures such as pre-flight prophylaxis, training, and possible therapeutics to reduce long-term effects. METHODOLOGY A literature search was performed for experimental studies conducted in astronauts and in animal models that simulated the space environment. Many studies, however, only discussed these with scientific reasoning and did not include any experimental methods. Relevant studies were identified through searching research databases such as PubMed and Google Scholar. No inclusion or exclusion criteria were used. FINDINGS Analysis of these studies provided a holistic understanding of the acute and chronic neurological changes that occur during space flight. Astronauts are exposed to hazards that include microgravity, cosmic radiation, hypercapnia, isolation, confinement and disrupted circadian rhythms. Microgravity, the absence of a gravitational force, is linked to disturbances in the vestibular system, intracranial and intraocular pressures. Furthermore, microgravity affects near field vision as part of the spaceflight-associated neuro-ocular syndrome. Exposure to cosmic radiation can increase the risk of neurodegenerative conditions and malignancies. It is estimated that cosmic radiation has significantly higher ionising capabilities than the ionising radiation used in medicine. Space travel also has potential benefits to the nervous system, including psychological development and effects on learning and memory. Future work needs to focus on how we can compare a current astronaut to a future space tourist. Potentially the physiological and psychological stresses of space flight might lead to neurological complications in future space travellers that do not have the physiological reserve of current astronauts.
Collapse
Affiliation(s)
- Udit Gupta
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385a Glossop Road, Sheffield and S10 2HQ, United Kingdom
| | - Sheharyar Baig
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385a Glossop Road, Sheffield and S10 2HQ, United Kingdom; Department of Clinical Neurology, Royal Hallamshire Hospital, Glossop Road, Sheffield, United Kingdom
| | - Arshad Majid
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385a Glossop Road, Sheffield and S10 2HQ, United Kingdom; Department of Clinical Neurology, Royal Hallamshire Hospital, Glossop Road, Sheffield, United Kingdom
| | - Simon M Bell
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385a Glossop Road, Sheffield and S10 2HQ, United Kingdom; Department of Clinical Neurology, Royal Hallamshire Hospital, Glossop Road, Sheffield, United Kingdom.
| |
Collapse
|
49
|
Huff JL, Poignant F, Rahmanian S, Khan N, Blakely EA, Britten RA, Chang P, Fornace AJ, Hada M, Kronenberg A, Norman RB, Patel ZS, Shay JW, Weil MM, Simonsen LC, Slaba TC. Galactic cosmic ray simulation at the NASA space radiation laboratory - Progress, challenges and recommendations on mixed-field effects. LIFE SCIENCES IN SPACE RESEARCH 2023; 36:90-104. [PMID: 36682835 DOI: 10.1016/j.lssr.2022.09.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 09/01/2022] [Accepted: 09/05/2022] [Indexed: 06/17/2023]
Abstract
For missions beyond low Earth orbit to the moon or Mars, space explorers will encounter a complex radiation field composed of various ion species with a broad range of energies. Such missions pose significant radiation protection challenges that need to be solved in order to minimize exposures and associated health risks. An innovative galactic cosmic ray simulator (GCRsim) was recently developed at the NASA Space Radiation Laboratory (NSRL) at Brookhaven National Laboratory (BNL). The GCRsim technology is intended to represent major components of the space radiation environment in a ground analog laboratory setting where it can be used to improve understanding of biological risks and serve as a testbed for countermeasure development and validation. The current GCRsim consists of 33 energetic ion beams that collectively simulate the primary and secondary GCR field encountered by humans in space over the broad range of particle types, energies, and linear energy transfer (LET) of interest to health effects. A virtual workshop was held in December 2020 to assess the status of the NASA baseline GCRsim. Workshop attendees examined various aspects of simulator design, with a particular emphasis on beam selection strategies. Experimental results, modeling approaches, areas of consensus, and questions of concern were also discussed in detail. This report includes a summary of the GCRsim workshop and a description of the current status of the GCRsim. This information is important for future advancements and applications in space radiobiology.
Collapse
Affiliation(s)
- Janice L Huff
- NASA Langley Research Center, Hampton, VA, 23681, United States of America.
| | - Floriane Poignant
- National Institute of Aerospace, Hampton, VA, 23666, United States of America
| | - Shirin Rahmanian
- National Institute of Aerospace, Hampton, VA, 23666, United States of America
| | - Nafisah Khan
- National Institute of Aerospace, Hampton, VA, 23666, United States of America
| | - Eleanor A Blakely
- Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, United States of America
| | - Richard A Britten
- Department of Radiation Oncology, Department of Microbiology and Molecular Cell Biology, Leroy T Canoles Jr. Cancer Center, School of Medicine, Eastern Virginia Medical School, Norfolk, VA, 23507, United States of America
| | - Polly Chang
- SRI International, Menlo Park, CA, 94025, United States of America
| | - Albert J Fornace
- Georgetown University, Washington, DC, 20057, United States of America
| | - Megumi Hada
- Prairie View A&M University, Prairie View, TX, 77446, United States of America
| | - Amy Kronenberg
- Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, United States of America
| | - Ryan B Norman
- NASA Langley Research Center, Hampton, VA, 23681, United States of America
| | - Zarana S Patel
- KBR Inc., Houston, TX, 77058, United States of America; NASA Johnson Space Center, Houston, TX, 77058, United States of America
| | - Jerry W Shay
- University of Texas Southwestern Medical Center, Dallas, TX, 75390, United States of America
| | - Michael M Weil
- Colorado State University, Fort Collins, CO, 80523, United States of America
| | - Lisa C Simonsen
- NASA Headquarters, Washington, DC, 20546, United States of America
| | - Tony C Slaba
- NASA Langley Research Center, Hampton, VA, 23681, United States of America
| |
Collapse
|
50
|
Ramos RL, Carante MP, Ferrari A, Sala P, Vercesi V, Ballarini F. A Mission to Mars: Prediction of GCR Doses and Comparison with Astronaut Dose Limits. Int J Mol Sci 2023; 24:2328. [PMID: 36768652 PMCID: PMC9916691 DOI: 10.3390/ijms24032328] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/17/2023] [Accepted: 01/19/2023] [Indexed: 01/26/2023] Open
Abstract
Long-term human space missions such as a future journey to Mars could be characterized by several hazards, among which radiation is one the highest-priority problems for astronaut health. In this work, exploiting a pre-existing interface between the BIANCA biophysical model and the FLUKA Monte Carlo transport code, a study was performed to calculate astronaut absorbed doses and equivalent doses following GCR exposure under different shielding conditions. More specifically, the interface with BIANCA allowed us to calculate both the RBE for cell survival, which is related to non-cancer effects, and that for chromosome aberrations, related to the induction of stochastic effects, including cancer. The results were then compared with cancer and non-cancer astronaut dose limits. Concerning the stochastic effects, the equivalent doses calculated by multiplying the absorbed dose by the RBE for chromosome aberrations ("high-dose method") were similar to those calculated using the Q-values recommended by ICRP. For a 650-day mission at solar minimum (representative of a possible Mars mission scenario), the obtained values are always lower than the career limit recommended by ICRP (1 Sv), but higher than the limit of 600 mSv recently adopted by NASA. The comparison with the JAXA limits is more complex, since they are age and sex dependent. Concerning the deterministic limits, even for a 650-day mission at solar minimum, the values obtained by multiplying the absorbed dose by the RBE for cell survival are largely below the limits established by the various space agencies. Following this work, BIANCA, interfaced with an MC transport code such as FLUKA, can now predict RBE values for cell death and chromosome aberrations following GCR exposure. More generally, both at solar minimum and at solar maximum, shielding of 10 g/cm2 Al seems to be a better choice than 20 g/cm2 for astronaut protection against GCR.
Collapse
Affiliation(s)
| | - Mario P. Carante
- INFN, Sezione di Pavia, Via Bassi 6, 27100 Pavia, Italy
- Physics Department, University of Pavia, Via Bassi 6, 27100 Pavia, Italy
| | - Alfredo Ferrari
- Institute for Astroparticle Physics, Karlsruhe Institute of Technology, 76021 Karlsruhe, Germany
| | - Paola Sala
- INFN, Sezione di Milano, Via Celoria 16, 20133 Milano, Italy
| | | | - Francesca Ballarini
- INFN, Sezione di Pavia, Via Bassi 6, 27100 Pavia, Italy
- Physics Department, University of Pavia, Via Bassi 6, 27100 Pavia, Italy
| |
Collapse
|