1
|
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
|
2
|
Seidler RD, Mao XW, Tays GD, Wang T, Zu Eulenburg P. Effects of spaceflight on the brain. Lancet Neurol 2024; 23:826-835. [PMID: 38945144 DOI: 10.1016/s1474-4422(24)00224-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 04/06/2024] [Accepted: 05/14/2024] [Indexed: 07/02/2024]
Abstract
The number of long duration human spaceflights has increased substantially over the past 15 years, leading to the discovery of numerous effects on the CNS. Microgravity results in headward fluid shifts, ventricular expansion, an upward shift of the brain within the skull, and remodelling of grey and white matter. The fluid changes are correlated with changes to perivascular space and spaceflight associated neuro-ocular syndrome. Microgravity alters the vestibular processing of head tilt and results in reduced tactile and proprioceptive inputs during spaceflight. Sensory adaptation is reflected in postflight effects, evident as transient sensorimotor impairment. Another major concern is that galactic cosmic radiation, which spacefarers will be exposed to when going beyond the magnetosphere around Earth, might have a negative effect on CNS function. Research with rodents points to the potential disruptive effects of space radiation on blood-brain barrier integrity and brain structures. More work is needed to understand and mitigate these effects on the CNS before humans travel to Mars, as the flight durations will be longer than anyone has previously experienced.
Collapse
Affiliation(s)
- Rachael D Seidler
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA; Department of Neurology, University of Florida, Gainesville, FL, USA.
| | - Xiao Wen Mao
- Department of Basic Sciences, Division of Biomedical Engineering Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Grant D Tays
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA
| | - Tianyi Wang
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA
| | - Peter Zu Eulenburg
- Institute for Neuroradiology, Ludwig-Maximilians University Munich, Munich, Germany
| |
Collapse
|
3
|
Strohm AO, Johnston C, Hernady E, Marples B, O'Banion MK, Majewska AK. Cranial irradiation disrupts homeostatic microglial dynamic behavior. J Neuroinflammation 2024; 21:82. [PMID: 38570852 PMCID: PMC10993621 DOI: 10.1186/s12974-024-03073-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: 12/18/2023] [Accepted: 03/22/2024] [Indexed: 04/05/2024] Open
Abstract
Cranial irradiation causes cognitive deficits that are in part mediated by microglia, the resident immune cells of the brain. Microglia are highly reactive, exhibiting changes in shape and morphology depending on the function they are performing. Additionally, microglia processes make dynamic, physical contacts with different components of their environment to monitor the functional state of the brain and promote plasticity. Though evidence suggests radiation perturbs homeostatic microglia functions, it is unknown how cranial irradiation impacts the dynamic behavior of microglia over time. Here, we paired in vivo two-photon microscopy with a transgenic mouse model that labels cortical microglia to follow these cells and determine how they change over time in cranial irradiated mice and their control littermates. We show that a single dose of 10 Gy cranial irradiation disrupts homeostatic cortical microglia dynamics during a 1-month time course. We found a lasting loss of microglial cells following cranial irradiation, coupled with a modest dysregulation of microglial soma displacement at earlier timepoints. The homogeneous distribution of microglia was maintained, suggesting microglia rearrange themselves to account for cell loss and maintain territorial organization following cranial irradiation. Furthermore, we found cranial irradiation reduced microglia coverage of the parenchyma and their surveillance capacity, without overtly changing morphology. Our results demonstrate that a single dose of radiation can induce changes in microglial behavior and function that could influence neurological health. These results set the foundation for future work examining how cranial irradiation impacts complex cellular dynamics in the brain which could contribute to the manifestation of cognitive deficits.
Collapse
Affiliation(s)
- Alexandra O Strohm
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Carl Johnston
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Eric Hernady
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Brian Marples
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - M Kerry O'Banion
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, 14642, USA
- Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Ania K Majewska
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, 14642, USA.
- Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, NY, 14642, USA.
- Center for Visual Science, University of Rochester Medical Center, Rochester, NY, 14642, USA.
| |
Collapse
|
4
|
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
|
5
|
Ye Z, Wang J, Shi W, Zhou Z, Zhang Y, Wang J, Yang H. Reprimo (RPRM) as a Potential Preventive and Therapeutic Target for Radiation-Induced Brain Injury via Multiple Mechanisms. Int J Mol Sci 2023; 24:17055. [PMID: 38069378 PMCID: PMC10707327 DOI: 10.3390/ijms242317055] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/09/2023] [Accepted: 11/23/2023] [Indexed: 12/18/2023] Open
Abstract
Patients receiving cranial radiotherapy for primary and metastatic brain tumors may experience radiation-induced brain injury (RIBI). Thus far, there has been a lack of effective preventive and therapeutic strategies for RIBI. Due to its complicated underlying pathogenic mechanisms, it is rather difficult to develop a single approach to target them simultaneously. We have recently reported that Reprimo (RPRM), a tumor suppressor gene, is a critical player in DNA damage repair, and RPRM deletion significantly confers radioresistance to mice. Herein, by using an RPRM knockout (KO) mouse model established in our laboratory, we found that RPRM deletion alleviated RIBI in mice via targeting its multiple underlying mechanisms. Specifically, RPRM knockout significantly reduced hippocampal DNA damage and apoptosis shortly after mice were exposed to whole-brain irradiation (WBI). For the late-delayed effect of WBI, RPRM knockout obviously ameliorated a radiation-induced decline in neurocognitive function and dramatically diminished WBI-induced neurogenesis inhibition. Moreover, RPRM KO mice exhibited a significantly lower level of acute and chronic inflammation response and microglial activation than wild-type (WT) mice post-WBI. Finally, we uncovered that RPRM knockout not only protected microglia against radiation-induced damage, thus preventing microglial activation, but also protected neurons and decreased the induction of CCL2 in neurons after irradiation, in turn attenuating the activation of microglial cells nearby through paracrine CCL2. Taken together, our results indicate that RPRM plays a crucial role in the occurrence of RIBI, suggesting that RPRM may serve as a novel potential target for the prevention and treatment of RIBI.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Hongying Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College of Soochow University, Suzhou 215123, China; (Z.Y.); (J.W.); (W.S.); (Z.Z.); (Y.Z.); (J.W.)
| |
Collapse
|
6
|
Desai RI, Kangas BD, Luc OT, Solakidou E, Smith EC, Dawes MH, Ma X, Makriyannis A, Chatterjee S, Dayeh MA, Muñoz-Jaramillo A, Desai MI, Limoli CL. Complex 33-beam simulated galactic cosmic radiation exposure impacts cognitive function and prefrontal cortex neurotransmitter networks in male mice. Nat Commun 2023; 14:7779. [PMID: 38012180 PMCID: PMC10682413 DOI: 10.1038/s41467-023-42173-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 09/28/2023] [Indexed: 11/29/2023] Open
Abstract
Astronauts will encounter extended exposure to galactic cosmic radiation (GCR) during deep space exploration, which could impair brain function. Here, we report that in male mice, acute or chronic GCR exposure did not modify reward sensitivity but did adversely affect attentional processes and increased reaction times. Potassium (K+)-stimulation in the prefrontal cortex (PFC) elevated dopamine (DA) but abolished temporal DA responsiveness after acute and chronic GCR exposure. Unlike acute GCR, chronic GCR increased levels of all other neurotransmitters, with differences evident between groups after higher K+-stimulation. Correlational and machine learning analysis showed that acute and chronic GCR exposure differentially reorganized the connection strength and causation of DA and other PFC neurotransmitter networks compared to controls which may explain space radiation-induced neurocognitive deficits.
Collapse
Affiliation(s)
- Rajeev I Desai
- Department of Psychiatry, Harvard Medical School, Boston, MA, 02115, USA.
- Behavioral Biology Program, McLean Hospital, Belmont, MA, 02478, USA.
- Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, 02115, USA.
| | - Brian D Kangas
- Department of Psychiatry, Harvard Medical School, Boston, MA, 02115, USA
- Behavioral Biology Program, McLean Hospital, Belmont, MA, 02478, USA
| | - Oanh T Luc
- Department of Psychiatry, Harvard Medical School, Boston, MA, 02115, USA
- Behavioral Biology Program, McLean Hospital, Belmont, MA, 02478, USA
| | - Eleana Solakidou
- Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, 02115, USA
- Medical School, University of Crete, Heraklion, Greece
| | - Evan C Smith
- Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, 02115, USA
| | - Monica H Dawes
- Department of Psychiatry, Harvard Medical School, Boston, MA, 02115, USA
- Behavioral Biology Program, McLean Hospital, Belmont, MA, 02478, USA
| | - Xiaoyu Ma
- Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, 02115, USA
| | - Alexandros Makriyannis
- Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, 02115, USA
| | | | - Maher A Dayeh
- Southwest Research Institute, San Antonio, TX, 78238, USA
- University of San Antonio, San Antonio, TX, 78249, USA
| | | | - Mihir I Desai
- Southwest Research Institute, San Antonio, TX, 78238, USA
- University of San Antonio, San Antonio, TX, 78249, USA
| | - Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine, Orange, CA, 92697, USA
| |
Collapse
|
7
|
Britten RA, Fesshaye A, Tidmore A, Liu A, Blackwell AA. Loss of Cognitive Flexibility Practice Effects in Female Rats Exposed to Simulated Space Radiation. Radiat Res 2023; 200:256-265. [PMID: 37527363 DOI: 10.1667/rade-22-00196.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 06/27/2023] [Indexed: 08/03/2023]
Abstract
During the planned missions to Mars, astronauts will be faced with many potential health hazards including prolonged exposure to space radiation. Ground-based studies have shown that exposure to space radiation impairs the performance of male rats in cognitive flexibility tasks which involve processes that are essential to rapidly and efficiently adapting to different situations. However, there is presently a paucity of information on the effects of space radiation on cognitive flexibility in female rodents. This study has determined the impact that exposure to a low (10 cGy) dose of ions from the simplified 5-ion galactic cosmic ray simulation [https://www.bnl.gov/nsrl/userguide/SimGCRSim.php (07/2023)] (GCRSim) beam or 250 MeV/n 4He ions has on the ability of female Wistar rats to perform in constrained [attentional set shifting (ATSET)] and unconstrained cognitive flexibility (UCFlex) tasks. Female rats exposed to GCRSim exhibited multiple decrements in ATSET performance. Firstly, GCRSim exposure impaired performance in the compound discrimination (CD) stage of the ATSET task. While the ability of rats to identify the rewarded cue was not compromised, the time the rats required to do so significantly increased. Secondly, both 4He and GCRSim exposure reduced the ability of rats to reach criterion in the compound discrimination reversal (CDR) stage. Approximately 20% of the irradiated rats were unable to complete the CDR task; furthermore, the irradiated rats that did reach criterion took more attempts to do so than did the sham-treated animals. Radiation exposure also altered the magnitude and/or nature of practice effects. A comparison of performance metrics from the pre-screen and post-exposure ATSET task revealed that while the sham-treated rats completed the post-exposure CD stage of the ATSET task in 30% less time than for completion of the pre-screen ATSET task, the irradiated rats took 30-50% longer to do so. Similarly, while sham-treated rats completed the CDR stage in ∼10% fewer attempts in the post-exposure task compared to the pre-screen task, in contrast, the 4He- and GCRSim-exposed cohorts took more (∼2-fold) attempts to reach criterion in the post-exposure task than in the pre-screen task. In conclusion, this study demonstrates that female rats are susceptible to radiation-induced loss of performance in the constrained ATSET cognitive flexibility task. Moreover, exposure to radiation leads to multiple performance decrements, including loss of practice effects, an increase in anterograde interference and reduced ability or unwillingness to switch attention. Should similar effects occur in humans, astronauts may have a compromised ability to perform complex tasks.
Collapse
Affiliation(s)
- Richard A Britten
- EVMS Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507
- EVMS Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, Virginia 23507
- Center for Integrative Neuroscience and Inflammatory diseases, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Arriyam Fesshaye
- EVMS Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Alyssa Tidmore
- EVMS Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Aiyi Liu
- EVMS Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Ashley A Blackwell
- EVMS Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507
- Center for Integrative Neuroscience and Inflammatory diseases, Eastern Virginia Medical School, Norfolk, Virginia 23507
| |
Collapse
|
8
|
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: 0] [Impact Index Per Article: 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
|
9
|
Allen BD, Alaghband Y, Kramár EA, Ru N, Petit B, Grilj V, Petronek MS, Pulliam CF, Kim RY, Doan NL, Baulch JE, Wood MA, Bailat C, Spitz DR, Vozenin MC, Limoli CL. Elucidating the neurological mechanism of the FLASH effect in juvenile mice exposed to hypofractionated radiotherapy. Neuro Oncol 2023; 25:927-939. [PMID: 36334265 PMCID: PMC10158064 DOI: 10.1093/neuonc/noac248] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Ultrahigh dose-rate radiotherapy (FLASH-RT) affords improvements in the therapeutic index by minimizing normal tissue toxicities without compromising antitumor efficacy compared to conventional dose-rate radiotherapy (CONV-RT). To investigate the translational potential of FLASH-RT to a human pediatric medulloblastoma brain tumor, we used a radiosensitive juvenile mouse model to assess adverse long-term neurological outcomes. METHODS Cohorts of 3-week-old male and female C57Bl/6 mice exposed to hypofractionated (2 × 10 Gy, FLASH-RT or CONV-RT) whole brain irradiation and unirradiated controls underwent behavioral testing to ascertain cognitive status four months posttreatment. Animals were sacrificed 6 months post-irradiation and tissues were analyzed for neurological and cerebrovascular decrements. RESULTS The neurological impact of FLASH-RT was analyzed over a 6-month follow-up. FLASH-RT ameliorated neurocognitive decrements induced by CONV-RT and preserved synaptic plasticity and integrity at the electrophysiological (long-term potentiation), molecular (synaptophysin), and structural (Bassoon/Homer-1 bouton) levels in multiple brain regions. The benefits of FLASH-RT were also linked to reduced neuroinflammation (activated microglia) and the preservation of the cerebrovascular structure, by maintaining aquaporin-4 levels and minimizing microglia colocalized to vessels. CONCLUSIONS Hypofractionated FLASH-RT affords significant and long-term normal tissue protection in the radiosensitive juvenile mouse brain when compared to CONV-RT. The capability of FLASH-RT to preserve critical cognitive outcomes and electrophysiological properties over 6-months is noteworthy and highlights its potential for resolving long-standing complications faced by pediatric brain tumor survivors. While care must be exercised before clinical translation is realized, present findings document the marked benefits of FLASH-RT that extend from synapse to cognition and the microvasculature.
Collapse
Affiliation(s)
- Barrett D Allen
- Department of Radiation Oncology, University of California, Irvine, CA 92697-2695, USA
| | - Yasaman Alaghband
- Department of Radiation Oncology, University of California, Irvine, CA 92697-2695, USA
| | - Eniko A Kramár
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Ning Ru
- Department of Radiation Oncology, University of California, Irvine, CA 92697-2695, USA
| | - Benoit Petit
- Laboratory of Radiation Oncology, Department of Radiation Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Veljko Grilj
- Laboratory of Radiation Oncology, Department of Radiation Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Michael S Petronek
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA 52242, USA
| | - Casey F Pulliam
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA 52242, USA
| | - Rachel Y Kim
- Department of Radiation Oncology, University of California, Irvine, CA 92697-2695, USA
| | - Ngoc-Lien Doan
- Department of Radiation Oncology, University of California, Irvine, CA 92697-2695, USA
| | - Janet E Baulch
- Department of Radiation Oncology, University of California, Irvine, CA 92697-2695, USA
| | - Marcelo A Wood
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Claude Bailat
- Institute of Radiation Physics/CHUV, Lausanne University Hospital, Lausanne, Switzerland
| | - Douglas R Spitz
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA 52242, USA
| | - Marie-Catherine Vozenin
- Laboratory of Radiation Oncology, Department of Radiation Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine, CA 92697-2695, USA
| |
Collapse
|
10
|
Usupzhanova DY, Astrelina TA, Kobzeva IV, Suchkova YB, Brunchukov VA, Rastorgueva AA, Nikitina VA, Samoilov AS. Evaluation of Changes in Some Functional Properties of Human Mesenchymal Stromal Cells Induced by Low Doses of Ionizing Radiation. Int J Mol Sci 2023; 24:6346. [PMID: 37047317 PMCID: PMC10094729 DOI: 10.3390/ijms24076346] [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: 12/15/2022] [Revised: 03/09/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
Each person is inevitably exposed to low doses of ionizing radiation (LDIR) throughout their life. The research results of LDIR effects are ambiguous and an accurate assessment of the risks associated with the influence of LDIR is an important task. Mesenchymal stromal cells (MSCs) are the regenerative reserve of an adult organism; because of this, they are a promising model for studying the effects of LDIR. The qualitative and quantitative changes in their characteristics can also be considered promising criteria for assessing the risks of LDIR exposure. The MSCs from human connective gingiva tissue (hG-MSCs) were irradiated at doses of 50, 100, 250, and 1000 mGy by the X-ray unit RUST-M1 (Russia). The cells were cultured continuously for 64 days after irradiation. During the study, we evaluated the secretory profile of hG-MSCs (IL-10, IDO, IL-6, IL-8, VEGF-A) using an ELISA test, the immunophenotype (CD45, CD34, CD90, CD105, CD73, HLA-DR, CD44) using flow cytometry, and the proliferative activity using the xCelligence RTCA cell analyzer at the chosen time points. The results of study have indicated the development of stimulating effects in the early stages of cultivation after irradiation using low doses of X-ray radiation. On the contrary, the effects of the low doses were comparable with the effects of medium doses of X-ray radiation in the long-term periods of cultivation after irradiation and have indicated the inhibition of the functional activity of MSCs.
Collapse
Affiliation(s)
| | - Tatiana A. Astrelina
- State Research Center-Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, 123098 Moscow, Russia
| | | | | | | | | | | | | |
Collapse
|
11
|
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
|
12
|
Nemec-Bakk AS, Sridharan V, Desai P, Landes RD, Hart B, Allen AR, Boerma M. Effects of Simulated 5-Ion Galactic Cosmic Radiation on Function and Structure of the Mouse Heart. Life (Basel) 2023; 13:life13030795. [PMID: 36983950 PMCID: PMC10057791 DOI: 10.3390/life13030795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/08/2023] [Accepted: 03/12/2023] [Indexed: 03/17/2023] Open
Abstract
Missions into deep space will expose astronauts to the harsh space environment, and the degenerative tissue effects of space radiation are largely unknown. To assess the risks, in this study, male BALB/c mice were exposed to 500 mGy 5-ion simulated GCR (GCRsim) at the NASA Space Radiation Laboratory. In addition, male and female CD1 mice were exposed to GCRsim and administered a diet containing Transforming Growth Factor-beta (TGF-β)RI kinase (ALK5) inhibitor IPW-5371 as a potential countermeasure. An ultrasound was performed to investigate cardiac function. Cardiac tissue was collected to determine collagen deposition, the density of the capillary network, and the expression of the immune mediator toll-like receptor 4 (TLR4) and immune cell markers CD2, CD4, and CD45. In male BALB/c mice, the only significant effects of GCRsim were an increase in the CD2 and TLR4 markers. In male CD1 mice, GCRsim caused a significant increase in total collagens and a decrease in the expression of TLR4, both of which were mitigated by the TGF-β inhibitor diet. In female CD1 mice, GCRsim caused an increase in the number of capillaries per tissue area in the ventricles, which may be explained by the decrease in the left ventricular mass. However, this increase was not mitigated by TGF-β inhibition. In both male and female CD1 mice, the combination of GCRsim and TGF-β inhibition caused changes in left ventricular immune cell markers that were not seen with GCRsim alone. These data suggest that GCRsim results in minor changes to cardiac tissue in both an inbred and outbred mouse strain. While there were few GCRsim effects to be mitigated, results from the combination of GCRsim and the TGF-β inhibitor do point to a role for TGF-β in maintaining markers of immune cells in the heart after exposure to GCR.
Collapse
Affiliation(s)
- Ashley S. Nemec-Bakk
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Correspondence:
| | - Vijayalakshmi Sridharan
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Parth Desai
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Reid D. Landes
- Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Barry Hart
- Innovation Pathways, LLC of Palo Alto, Palo Alto, CA 94301, USA
| | - Antiño R. Allen
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Marjan Boerma
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
13
|
Alaghband Y, Klein PM, Kramár EA, Cranston MN, Perry BC, Shelerud LM, Kane AE, Doan NL, Ru N, Acharya MM, Wood MA, Sinclair DA, Dickstein DL, Soltesz I, Limoli CL, Baulch JE. Galactic cosmic radiation exposure causes multifaceted neurocognitive impairments. Cell Mol Life Sci 2023; 80:29. [PMID: 36607431 PMCID: PMC9823026 DOI: 10.1007/s00018-022-04666-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 11/01/2022] [Accepted: 12/11/2022] [Indexed: 01/07/2023]
Abstract
Technological advancements have facilitated the implementation of realistic, terrestrial-based complex 33-beam galactic cosmic radiation simulations (GCR Sim) to now probe central nervous system functionality. This work expands considerably on prior, simplified GCR simulations, yielding new insights into responses of male and female mice exposed to 40-50 cGy acute or chronic radiations relevant to deep space travel. Results of the object in updated location task suggested that exposure to acute or chronic GCR Sim induced persistent impairments in hippocampus-dependent memory formation and reconsolidation in female mice that did not manifest robustly in irradiated male mice. Interestingly, irradiated male mice, but not females, were impaired in novel object recognition and chronically irradiated males exhibited increased aggressive behavior on the tube dominance test. Electrophysiology studies used to evaluate synaptic plasticity in the hippocampal CA1 region revealed significant reductions in long-term potentiation after each irradiation paradigm in both sexes. Interestingly, network-level disruptions did not translate to altered intrinsic electrophysiological properties of CA1 pyramidal cells, whereas acute exposures caused modest drops in excitatory synaptic signaling in males. Ultrastructural analyses of CA1 synapses found smaller postsynaptic densities in larger spines of chronically exposed mice compared to controls and acutely exposed mice. Myelination was also affected by GCR Sim with acutely exposed mice exhibiting an increase in the percent of myelinated axons; however, the myelin sheathes on small calibur (< 0.3 mm) and larger (> 0.5 mm) axons were thinner when compared to controls. Present findings might have been predicted based on previous studies using single and mixed beam exposures and provide further evidence that space-relevant radiation exposures disrupt critical cognitive processes and underlying neuronal network-level plasticity, albeit not to the extent that might have been previously predicted.
Collapse
Affiliation(s)
- Yasaman Alaghband
- Department of Radiation Oncology, Medical Sciences I, University of California Irvine, Room B-146D, Irvine, CA, 92697-2695, USA
| | - Peter M Klein
- Department of Neurosurgery, Stanford University, Palo Alto, CA, 94305, USA
| | - Eniko A Kramár
- Department of Neurobiology and Behavior, School of Biological Sciences, University of California, Irvine, 92697-2695, USA
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, 92697-2695, USA
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, 92697-2695, USA
| | - Michael N Cranston
- Department of Pathology, Uniformed Services University of Health Sciences, Bethesda, MD, 20814, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine (HJF), Bethesda, MD, 20817, USA
| | - Bayley C Perry
- Department of Pathology, Uniformed Services University of Health Sciences, Bethesda, MD, 20814, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine (HJF), Bethesda, MD, 20817, USA
| | - Lukas M Shelerud
- Department of Genetics, Blavatnik Institute, Paul F. Glenn Center for Biology of Aging Research, Harvard Medical School, Boston, MA, 0211, USA
| | - Alice E Kane
- Department of Genetics, Blavatnik Institute, Paul F. Glenn Center for Biology of Aging Research, Harvard Medical School, Boston, MA, 0211, USA
| | - Ngoc-Lien Doan
- Department of Radiation Oncology, Medical Sciences I, University of California Irvine, Room B-146D, Irvine, CA, 92697-2695, USA
| | - Ning Ru
- Department of Radiation Oncology, Medical Sciences I, University of California Irvine, Room B-146D, Irvine, CA, 92697-2695, USA
| | - Munjal M Acharya
- Department of Radiation Oncology, Medical Sciences I, University of California Irvine, Room B-146D, Irvine, CA, 92697-2695, USA
- Department of Anatomy and Neurobiology, University of California, Irvine, 92697-2695, USA
| | - Marcelo A Wood
- Department of Neurobiology and Behavior, School of Biological Sciences, University of California, Irvine, 92697-2695, USA
| | - David A Sinclair
- Department of Genetics, Blavatnik Institute, Paul F. Glenn Center for Biology of Aging Research, Harvard Medical School, Boston, MA, 0211, USA
| | - Dara L Dickstein
- Department of Pathology, Uniformed Services University of Health Sciences, Bethesda, MD, 20814, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine (HJF), Bethesda, MD, 20817, USA
| | - Ivan Soltesz
- Department of Neurosurgery, Stanford University, Palo Alto, CA, 94305, USA
- Department of Neurology and Neurological Sciences, Stanford University, Palo Alto, CA, 94305, USA
| | - Charles L Limoli
- Department of Radiation Oncology, Medical Sciences I, University of California Irvine, Room B-146D, Irvine, CA, 92697-2695, USA
| | - Janet E Baulch
- Department of Radiation Oncology, Medical Sciences I, University of California Irvine, Room B-146D, Irvine, CA, 92697-2695, USA.
| |
Collapse
|
14
|
Simmons P, Trujillo M, McElroy T, Binz R, Pathak R, Allen AR. Evaluating the effects of low-dose simulated galactic cosmic rays on murine hippocampal-dependent cognitive performance. Front Neurosci 2022; 16:908632. [PMID: 36561122 PMCID: PMC9765097 DOI: 10.3389/fnins.2022.908632] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 09/12/2022] [Indexed: 12/12/2022] Open
Abstract
Space exploration has advanced substantially over recent decades and plans to increase the duration of deep space missions are in preparation. One of the primary health concerns is potential damage to the central nervous system (CNS), resulting in loss of cognitive abilities and function. The majority of ground-based research on space radiation-induced health risks has been conducted using single particle simulations, which do not effectively model real-world scenarios. Thus, to improve the safety of space missions, we must expand our understanding of the effects of simulated galactic cosmic rays (GCRs) on the CNS. To assess the effects of low-dose GCR, we subjected 6-month-old male BALB/c mice to 50 cGy 5-beam simplified GCR spectrum (1H, 28Si, 4He, 16O, and 56Fe) whole-body irradiation at the NASA Space Radiation Laboratory. Animals were tested for cognitive performance with Y-maze and Morris water maze tests 3 months after irradiation. Irradiated animals had impaired short-term memory and lacked spatial memory retention on day 5 of the probe trial. Glial cell analysis by flow cytometry showed no significant changes in oligodendrocytes, astrocytes, microglia or neural precursor cells (NPC's) between the sham group and GCR group. Bone marrow cytogenetic data showed a significant increase in the frequency of chromosomal aberrations after GCR exposure. Finally, tandem mass tag proteomics identified 3,639 proteins, 113 of which were differentially expressed when comparing sham versus GCR exposure (fold change > 1.5; p < 0.05). Our data suggest exposure to low-dose GCR induces cognitive deficits by impairing short-term memory and spatial memory retention.
Collapse
Affiliation(s)
- Pilar Simmons
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States,Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Madison Trujillo
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States,Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Taylor McElroy
- Department of Aging, University of Florida, Gainesville, FL, United States
| | - Regina Binz
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Rupak Pathak
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Antiño R. Allen
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States,Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States,*Correspondence: Antiño R. Allen,
| |
Collapse
|
15
|
Zablotska LB, Zupunski L, Leuraud K, Lopes J, Hinkle J, Pugeda T, Delgado T, Olschowka J, Williams J, O’Banion MK, Boice JD, Cohen SS, Mumma MT, Dauer LT, Britten RA, Stephenson S. Radiation and CNS effects: summary of evidence from a recent symposium of the Radiation Research Society. Int J Radiat Biol 2022; 99:1332-1342. [PMID: 36318723 PMCID: PMC10759179 DOI: 10.1080/09553002.2023.2142984] [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: 07/22/2022] [Revised: 09/26/2022] [Accepted: 09/28/2022] [Indexed: 11/13/2022]
Abstract
This article summarizes a Symposium on 'Radiation risks of the central nervous system' held virtually at the 67th Annual Meeting of the Radiation Research Society, 3-6 October 2021. Repeated low-dose radiation exposure over a certain period could lead to reduced neuronal proliferation, altered neurogenesis, neuroinflammation and various neurological complications, including psychological consequences, necessitating further research in these areas. Four speakers from radiation biology, genetics and epidemiology presented the latest data from their studies seeking insights into this important topic. This symposium highlighted new and important directions for further research on mental health disorders, neurodegenerative conditions and cognitive impairment. Future studies will examine risks of mental and behavioral disorders and neurodegenerative diseases following protracted radiation exposures to better understand risks of occupational exposures as well as provide insights into risks from exposures to galactic cosmic rays.
Collapse
Affiliation(s)
- Lydia B. Zablotska
- Department of Epidemiology and Biostatistics, School of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Ljubica Zupunski
- Environment and Lifestyle Epidemiology Branch, International Agency for Research on Cancer, Lyon, France
| | - Klervi Leuraud
- Laboratory of Epidemiology (LEPID), Institute for Radiological Protection and Nuclear Safety (IRSN), Fontenay-aux-Roses, France
| | - Julie Lopes
- Laboratory of Epidemiology (LEPID), Institute for Radiological Protection and Nuclear Safety (IRSN), Fontenay-aux-Roses, France
| | - Joshua Hinkle
- Department of Neuroscience, University of Rochester School of Medicine & Dentistry, Rochester, NY, USA
- Del Monte Institute for Neuroscience, University of Rochester School of Medicine & Dentistry, Rochester, NY, USA
| | - Tyler Pugeda
- Department of Neuroscience, University of Rochester School of Medicine & Dentistry, Rochester, NY, USA
- Del Monte Institute for Neuroscience, University of Rochester School of Medicine & Dentistry, Rochester, NY, USA
| | - Thomas Delgado
- Department of Neuroscience, University of Rochester School of Medicine & Dentistry, Rochester, NY, USA
- Del Monte Institute for Neuroscience, University of Rochester School of Medicine & Dentistry, Rochester, NY, USA
| | - John Olschowka
- Department of Neuroscience, University of Rochester School of Medicine & Dentistry, Rochester, NY, USA
- Del Monte Institute for Neuroscience, University of Rochester School of Medicine & Dentistry, Rochester, NY, USA
| | - Jacqueline Williams
- Department of Environmental Medicine, University of Rochester School of Medicine & Dentistry, Rochester, NY, USA
- Wilmot Cancer Center, University of Rochester School of Medicine & Dentistry, Rochester, NY, USA
| | - M. Kerry O’Banion
- Department of Neuroscience, University of Rochester School of Medicine & Dentistry, Rochester, NY, USA
- Del Monte Institute for Neuroscience, University of Rochester School of Medicine & Dentistry, Rochester, NY, USA
- Wilmot Cancer Center, University of Rochester School of Medicine & Dentistry, Rochester, NY, USA
- Department of Neurology, University of Rochester School of Medicine & Dentistry, Rochester, NY, USA
| | - John D. Boice
- National Council on Radiation Protection and Measurements, Bethesda, MD, USA
- Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sarah S. Cohen
- Vanderbilt University Medical Center, Nashville, TN, USA
- EpidStrategies, Cary, NC, USA
| | - Michael T. Mumma
- Vanderbilt University Medical Center, Nashville, TN, USA
- International Epidemiology Institute, Rockville, MD, USA
| | | | - Richard A. Britten
- Department of Radiation Oncology, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Samuel Stephenson
- School of Medicine, Eastern Virginia Medical School, Norfolk, VA, USA
| |
Collapse
|
16
|
Kerry O'Banion M. Microglia: Rheostats of space radiation effects in the CNS microenvironment. LIFE SCIENCES IN SPACE RESEARCH 2022; 35:180-186. [PMID: 36336364 DOI: 10.1016/j.lssr.2022.08.002] [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: 04/04/2022] [Revised: 08/01/2022] [Accepted: 08/04/2022] [Indexed: 06/16/2023]
Abstract
Microglia are innate immune cells within the brain that arise from a distinct myeloid lineage. Like other tissue resident macrophages, microglia respond to injury or immune challenges and participate in reparative processes such as phagocytosis to preserve normal function. Importantly, they also participate in normal homeostatic processes including maintenance of neurogenic niches and synaptic plasticity associated with development. This review highlights aspects of microglial biology and how repeated insults that occur with age, neurodegenerative disease and possibly radiation exposure may heighten microglial responses and contribute to their dysfunction, creating a situation where their normal reparative mechanisms are no longer sufficient to maintain brain health. These ideas are discussed in the context of an evolving literature focused on microglial responses as possible targets for mitigation of late CNS radiation effects that represent potential risks for future exploration of deep space environments.
Collapse
Affiliation(s)
- M Kerry O'Banion
- Department of Neuroscience, USA; Del Monte Institute for Neuroscience, USA; Wilmot Cancer Institute, University of Rochester School of Medicine & Dentistry, Rochester, NY 14642, USA.
| |
Collapse
|
17
|
Miller KB, Mi KL, Nelson GA, Norman RB, Patel ZS, Huff JL. Ionizing radiation, cerebrovascular disease, and consequent dementia: A review and proposed framework relevant to space radiation exposure. Front Physiol 2022; 13:1008640. [PMID: 36388106 PMCID: PMC9640983 DOI: 10.3389/fphys.2022.1008640] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 09/29/2022] [Indexed: 09/05/2023] Open
Abstract
Space exploration requires the characterization and management or mitigation of a variety of human health risks. Exposure to space radiation is one of the main health concerns because it has the potential to increase the risk of cancer, cardiovascular disease, and both acute and late neurodegeneration. Space radiation-induced decrements to the vascular system may impact the risk for cerebrovascular disease and consequent dementia. These risks may be independent or synergistic with direct damage to central nervous system tissues. The purpose of this work is to review epidemiological and experimental data regarding the impact of low-to-moderate dose ionizing radiation on the central nervous system and the cerebrovascular system. A proposed framework outlines how space radiation-induced effects on the vasculature may increase risk for both cerebrovascular dysfunction and neural and cognitive adverse outcomes. The results of this work suggest that there are multiple processes by which ionizing radiation exposure may impact cerebrovascular function including increases in oxidative stress, neuroinflammation, endothelial cell dysfunction, arterial stiffening, atherosclerosis, and cerebral amyloid angiopathy. Cerebrovascular adverse outcomes may also promote neural and cognitive adverse outcomes. However, there are many gaps in both the human and preclinical evidence base regarding the long-term impact of ionizing radiation exposure on brain health due to heterogeneity in both exposures and outcomes. The unique composition of the space radiation environment makes the translation of the evidence base from terrestrial exposures to space exposures difficult. Additional investigation and understanding of the impact of low-to-moderate doses of ionizing radiation including high (H) atomic number (Z) and energy (E) (HZE) ions on the cerebrovascular system is needed. Furthermore, investigation of how decrements in vascular systems may contribute to development of neurodegenerative diseases in independent or synergistic pathways is important for protecting the long-term health of astronauts.
Collapse
Affiliation(s)
| | | | - Gregory A. Nelson
- Department of Basic Sciences, Division of Biomedical Engineering Sciences, Loma Linda University, Loma Linda, CA, United States
- NASA Johnson Space Center, Houston, TX, United States
- KBR Inc., Houston, TX, United States
| | - Ryan B. Norman
- NASA Langley Research Center, Hampton, VA, United States
| | - Zarana S. Patel
- NASA Johnson Space Center, Houston, TX, United States
- KBR Inc., Houston, TX, United States
| | - Janice L. Huff
- NASA Langley Research Center, Hampton, VA, United States
| |
Collapse
|
18
|
Liu Q, Huang Y, Duan M, Yang Q, Ren B, Tang F. Microglia as Therapeutic Target for Radiation-Induced Brain Injury. Int J Mol Sci 2022; 23:8286. [PMID: 35955439 PMCID: PMC9368164 DOI: 10.3390/ijms23158286] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 12/10/2022] Open
Abstract
Radiation-induced brain injury (RIBI) after radiotherapy has become an increasingly important factor affecting the prognosis of patients with head and neck tumor. With the delivery of high doses of radiation to brain tissue, microglia rapidly transit to a pro-inflammatory phenotype, upregulate phagocytic machinery, and reduce the release of neurotrophic factors. Persistently activated microglia mediate the progression of chronic neuroinflammation, which may inhibit brain neurogenesis leading to the occurrence of neurocognitive disorders at the advanced stage of RIBI. Fully understanding the microglial pathophysiology and cellular and molecular mechanisms after irradiation may facilitate the development of novel therapy by targeting microglia to prevent RIBI and subsequent neurological and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Qun Liu
- The School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China; (Q.L.); (Y.H.)
| | - Yan Huang
- The School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China; (Q.L.); (Y.H.)
| | - Mengyun Duan
- Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou 434023, China; (M.D.); (Q.Y.)
| | - Qun Yang
- Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou 434023, China; (M.D.); (Q.Y.)
| | - Boxu Ren
- The School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China; (Q.L.); (Y.H.)
| | - Fengru Tang
- Radiation Physiology Laboratory, Singapore Nuclear Research and Safety Initiative, National University of Singapore, Singapore 138602, Singapore
| |
Collapse
|
19
|
Britten RA, Fesshaye A, Tidmore A, Blackwell AA. Similar Loss of Executive Function Performance after Exposure to Low (10 cGy) Doses of Single (4He) Ions and the Multi-Ion GCRSim Beam. Radiat Res 2022; 198:375-383. [DOI: 10.1667/rade-22-00022.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 06/14/2022] [Indexed: 11/03/2022]
|
20
|
Schaeffer EA, Blackwell AA, Oltmanns JRO, Einhaus R, Lake R, Hein CP, Baulch JE, Limoli CL, Ton ST, Kartje GL, Wallace DG. Differential organization of open field behavior in mice following acute or chronic simulated GCR exposure. Behav Brain Res 2022; 416:113577. [PMID: 34506841 DOI: 10.1016/j.bbr.2021.113577] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 08/27/2021] [Accepted: 09/04/2021] [Indexed: 11/20/2022]
Abstract
Astronauts undertaking deep space travel will receive chronic exposure to the mixed spectrum of particles that comprise Galactic Cosmic Radiation (GCR). Exposure to the different charged particles of varied fluence and energy that characterize GCR may impact neural systems that support performance on mission critical tasks. Indeed, growing evidence derived from years of terrestrial-based simulations of the space radiation environment using rodents has indicated that a variety of exposure scenarios can result in significant and long-lasting decrements to CNS functionality. Many of the behavioral tasks used to quantify radiation effects on the CNS depend on neural systems that support maintaining spatial orientation and organization of rodent open field behavior. The current study examined the effects of acute or chronic exposure to simulated GCR on the organization of open field behavior under conditions with varied access to environmental cues in male and female C57BL/6 J mice. In general, groups exhibited similar organization of open field behavior under dark and light conditions. Two exceptions were noted: the acute exposure group exhibited significantly slower and more circuitous homeward progressions relative to the chronic group under light conditions. These results demonstrate the potential of open field behavior organization to discriminate between the effects of select GCR exposure paradigms.
Collapse
Affiliation(s)
- E A Schaeffer
- Department of Psychology, Northern Illinois University, DeKalb, IL 60115, USA
| | - A A Blackwell
- Department of Psychology, Northern Illinois University, DeKalb, IL 60115, USA
| | | | - R Einhaus
- Department of Psychology, Northern Illinois University, DeKalb, IL 60115, USA
| | - R Lake
- Department of Psychology, Northern Illinois University, DeKalb, IL 60115, USA
| | - C Piwowar Hein
- Department of Psychology, Northern Illinois University, DeKalb, IL 60115, USA
| | - J E Baulch
- Department of Radiation Oncology, University of California Irvine, Irvine, CA, USA
| | - C L Limoli
- Department of Radiation Oncology, University of California Irvine, Irvine, CA, USA
| | - S T Ton
- Loyola University Health Sciences Division, Maywood, IL, USA; Edward Hines Jr. Veterans Affairs Hospital, Research Service, Hines, IL, USA
| | - G L Kartje
- Loyola University Health Sciences Division, Maywood, IL, USA; Edward Hines Jr. Veterans Affairs Hospital, Research Service, Hines, IL, USA
| | - D G Wallace
- Department of Psychology, Northern Illinois University, DeKalb, IL 60115, USA.
| |
Collapse
|
21
|
Long-Term Sex- and Genotype-Specific Effects of 56Fe Irradiation on Wild-Type and APPswe/PS1dE9 Transgenic Mice. Int J Mol Sci 2021; 22:ijms222413305. [PMID: 34948098 PMCID: PMC8703695 DOI: 10.3390/ijms222413305] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/07/2021] [Accepted: 12/07/2021] [Indexed: 12/22/2022] Open
Abstract
Space radiation presents a substantial threat to travel beyond Earth. Relatively low doses of high-energy particle radiation cause physiological and behavioral impairments in rodents and may pose risks to human spaceflight. There is evidence that 56Fe irradiation, a significant component of space radiation, may be more harmful to males than to females and worsen Alzheimer's disease pathology in genetically vulnerable models. Yet, research on the long-term, sex- and genotype-specific effects of 56Fe irradiation is lacking. Here, we irradiated 4-month-old male and female, wild-type and Alzheimer's-like APP/PS1 mice with 0, 0.10, or 0.50 Gy of 56Fe ions (1GeV/u). Mice underwent microPET scans before and 7.5 months after irradiation, a battery of behavioral tests at 11 months of age and were sacrificed for pathological and biochemical analyses at 12 months of age. 56Fe irradiation worsened amyloid-beta (Aβ) pathology, gliosis, neuroinflammation and spatial memory, but improved motor coordination, in male transgenic mice and worsened fear memory in wild-type males. Although sham-irradiated female APP/PS1 mice had more cerebral Aβ and gliosis than sham-irradiated male transgenics, female mice of both genotypes were relatively spared from radiation effects 8 months later. These results provide evidence for sex-specific, long-term CNS effects of space radiation.
Collapse
|
22
|
Boutros SW, Zimmerman B, Nagy SC, Lee JS, Perez R, Raber J. Amifostine (WR-2721) Mitigates Cognitive Injury Induced by Heavy Ion Radiation in Male Mice and Alters Behavior and Brain Connectivity. Front Physiol 2021; 12:770502. [PMID: 34867479 PMCID: PMC8637850 DOI: 10.3389/fphys.2021.770502] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 10/22/2021] [Indexed: 12/11/2022] Open
Abstract
The deep space environment contains many risks to astronauts during space missions, such as galactic cosmic rays (GCRs) comprised of naturally occurring heavy ions. Heavy ion radiation is increasingly being used in cancer therapy, including novel regimens involving carbon therapy. Previous investigations involving simulated space radiation have indicated a host of detrimental cognitive and behavioral effects. Therefore, there is an increasing need to counteract these deleterious effects of heavy ion radiation. Here, we assessed the ability of amifostine to mitigate cognitive injury induced by simulated GCRs in C57Bl/6J male and female mice. Six-month-old mice received an intraperitoneal injection of saline, 107 mg/kg, or 214 mg/kg of amifostine 1 h prior to exposure to a simplified five-ion radiation (protons, 28Si, 4He, 16O, and 56Fe) at 500 mGy or sham radiation. Mice were behaviorally tested 2-3 months later. Male mice that received saline and radiation exposure failed to show novel object recognition, which was reversed by both doses of amifostine. Conversely, female mice that received saline and radiation exposure displayed intact object recognition, but those that received amifostine prior to radiation did not. Amifostine and radiation also had distinct effects on males and females in the open field, with amifostine affecting distance moved over time in both sexes, and radiation affecting time spent in the center in females only. Whole-brain analysis of cFos immunoreactivity in male mice indicated that amifostine and radiation altered regional connectivity in areas involved in novel object recognition. These data support that amifostine has potential as a countermeasure against cognitive injury following proton and heavy ion irradiation in males.
Collapse
Affiliation(s)
- Sydney Weber Boutros
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Benjamin Zimmerman
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR, United States
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Sydney C. Nagy
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Joanne S. Lee
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Ruby Perez
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
- Departments of Neurology and Radiation Medicine, Oregon Health & Science University, Portland, OR, United States
- Division of Neuroscience, Oregon National Primate Research Center, Portland, OR, United States
| |
Collapse
|
23
|
Prelich MT, Matar M, Gokoglu SA, Gallo CA, Schepelmann A, Iqbal AK, Lewandowski BE, Britten RA, Prabhu RK, Myers JG. Predicting Space Radiation Single Ion Exposure in Rodents: A Machine Learning Approach. Front Syst Neurosci 2021; 15:715433. [PMID: 34720896 PMCID: PMC8555470 DOI: 10.3389/fnsys.2021.715433] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 09/21/2021] [Indexed: 11/13/2022] Open
Abstract
This study presents a data-driven machine learning approach to predict individual Galactic Cosmic Radiation (GCR) ion exposure for 4He, 16O, 28Si, 48Ti, or 56Fe up to 150 mGy, based on Attentional Set-shifting (ATSET) experimental tests. The ATSET assay consists of a series of cognitive performance tasks on irradiated male Wistar rats. The GCR ion doses represent the expected cumulative radiation astronauts may receive during a Mars mission on an individual ion basis. The primary objective is to synthesize and assess predictive models on a per-subject level through Machine Learning (ML) classifiers. The raw cognitive performance data from individual rodent subjects are used as features to train the models and to explore the capabilities of three different ML techniques for elucidating a range of correlations between received radiation on rodents and their performance outcomes. The analysis employs scores of selected input features and different normalization approaches which yield varying degrees of model performance. The current study shows that support vector machine, Gaussian naive Bayes, and random forest models are capable of predicting individual ion exposure using ATSET scores where corresponding Matthews correlation coefficients and F1 scores reflect model performance exceeding random chance. The study suggests a decremental effect on cognitive performance in rodents due to ≤150 mGy of single ion exposure, inasmuch as the models can discriminate between 0 mGy and any exposure level in the performance score feature space. A number of observations about the utility and limitations in specific normalization routines and evaluation scores are examined as well as best practices for ML with imbalanced datasets observed.
Collapse
Affiliation(s)
| | - Mona Matar
- NASA Glenn Research Center, Cleveland, OH, United States
| | | | | | | | - Asad K Iqbal
- ZIN Technologies, Inc., Cleveland, OH, United States
| | | | - Richard A Britten
- Department of Radiation Oncology, Eastern Virginia Medical School, Norfolk, VA, United States
| | - R K Prabhu
- Universities Space Research Association, Cleveland, OH, United States
| | - Jerry G Myers
- NASA Glenn Research Center, Cleveland, OH, United States
| |
Collapse
|
24
|
Cucinotta FA, Schimmerling W, Blakely EA, Hei TK. A proposed change to astronaut exposures limits is a giant leap backwards for radiation protection. LIFE SCIENCES IN SPACE RESEARCH 2021; 31:59-70. [PMID: 34689951 DOI: 10.1016/j.lssr.2021.07.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 07/24/2021] [Accepted: 07/25/2021] [Indexed: 06/13/2023]
Abstract
Addressing the uncertainties in assessing health risks from cosmic ray heavy ions is a major scientific challenge recognized by many previous reports by the National Academy of Sciences (NAS) and the National Council on Radiation Protection and Measurements (NCRP) advising the National Aeronautics and Space Administration (NASA). These reports suggested a series of steps to pursue the scientific basis for space radiation protection, including the implementation of age and sex dependent risk assessments and exposure limits appropriate for a small population of radiation workers, the evaluation of uncertainties in risk projections, and developing a vigorous research program in heavy ion radiobiology to reduce uncertainties and discover effective countermeasures. The assessment of uncertainties in assessing risk provides protection against changing assessments of risk, reveals limitations in information used in space mission operations, and provides the impetus to reduce uncertainties and discover the true level of risk and possible effectiveness of countermeasures through research. However, recommendations of a recent NAS report, in an effort to minimize differences in age and sex on flight opportunities, suggest a 600 mSv career effective dose limit based on a median estimate to reach 3% cancer fatality for 35-year old females. The NAS report does not call out examples where females would be excluded from space missions planned in the current decade using the current radiation limits at NASA. In addition, there are minimal considerations of the level of risk to be encountered at this exposure level with respect to the uncertainties of heavy ion radiobiology, and risks of cancer, as well as cognitive detriments and circulatory diseases. Furthermore, their recommendation to limit Sieverts and not risk in conjunction with a waiver process is essentially a recommendation to remove radiation limits for astronauts. We discuss issues with several of the NAS recommendations with the conclusion that the recommendations could have negative impacts on crew health and safety, and violate the three principles of radiation protection (to prevent clinically significant deterministic effects, limit stochastic effects, and practice ALARA), which would be a giant leap backwards for radiation protection.
Collapse
Affiliation(s)
- Francis A Cucinotta
- Department of Health Physics and Diagnostic Sciences, University of Nevada Las Vegas, Las Vegas, NV, USA.
| | | | | | - Tom K Hei
- Center for Radiological Research, Columbia University, New York, NY, USA
| |
Collapse
|
25
|
Soler I, Yun S, Reynolds RP, Whoolery CW, Tran FH, Kumar PL, Rong Y, DeSalle MJ, Gibson AD, Stowe AM, Kiffer FC, Eisch AJ. Multi-Domain Touchscreen-Based Cognitive Assessment of C57BL/6J Female Mice Shows Whole-Body Exposure to 56Fe Particle Space Radiation in Maturity Improves Discrimination Learning Yet Impairs Stimulus-Response Rule-Based Habit Learning. Front Behav Neurosci 2021; 15:722780. [PMID: 34707486 PMCID: PMC8543003 DOI: 10.3389/fnbeh.2021.722780] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 09/08/2021] [Indexed: 12/23/2022] Open
Abstract
Astronauts during interplanetary missions will be exposed to galactic cosmic radiation, including charged particles like 56Fe. Most preclinical studies with mature, "astronaut-aged" rodents suggest space radiation diminishes performance in classical hippocampal- and prefrontal cortex-dependent tasks. However, a rodent cognitive touchscreen battery unexpectedly revealed 56Fe radiation improves the performance of C57BL/6J male mice in a hippocampal-dependent task (discrimination learning) without changing performance in a striatal-dependent task (rule-based learning). As there are conflicting results on whether the female rodent brain is preferentially injured by or resistant to charged particle exposure, and as the proportion of female vs. male astronauts is increasing, further study on how charged particles influence the touchscreen cognitive performance of female mice is warranted. We hypothesized that, similar to mature male mice, mature female C57BL/6J mice exposed to fractionated whole-body 56Fe irradiation (3 × 6.7cGy 56Fe over 5 days, 600 MeV/n) would improve performance vs. Sham conditions in touchscreen tasks relevant to hippocampal and prefrontal cortical function [e.g., location discrimination reversal (LDR) and extinction, respectively]. In LDR, 56Fe female mice more accurately discriminated two discrete conditioned stimuli relative to Sham mice, suggesting improved hippocampal function. However, 56Fe and Sham female mice acquired a new simple stimulus-response behavior and extinguished this acquired behavior at similar rates, suggesting similar prefrontal cortical function. Based on prior work on multiple memory systems, we next tested whether improved hippocampal-dependent function (discrimination learning) came at the expense of striatal stimulus-response rule-based habit learning (visuomotor conditional learning). Interestingly, 56Fe female mice took more days to reach criteria in this striatal-dependent rule-based test relative to Sham mice. Together, our data support the idea of competition between memory systems, as an 56Fe-induced decrease in striatal-based learning is associated with enhanced hippocampal-based learning. These data emphasize the power of using a touchscreen-based battery to advance our understanding of the effects of space radiation on mission critical cognitive function in females, and underscore the importance of preclinical space radiation risk studies measuring multiple cognitive processes, thereby preventing NASA's risk assessments from being based on a single cognitive domain.
Collapse
Affiliation(s)
- Ivan Soler
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Sanghee Yun
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Ryan P. Reynolds
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Cody W. Whoolery
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Fionya H. Tran
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Priya L. Kumar
- University of Pennsylvania, Philadelphia, PA, United States
| | - Yuying Rong
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Matthew J. DeSalle
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Adam D. Gibson
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Ann M. Stowe
- Department of Neurology and Neurological Therapeutics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Frederico C. Kiffer
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Amelia J. Eisch
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Neuroscience, Perelman School of Medicine, Mahoney Institute for Neurosciences, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
26
|
Tidmore A, Dutta SM, Fesshaye AS, Russell WK, Duncan VD, Britten RA. Space Radiation-Induced Alterations in the Hippocampal Ubiquitin-Proteome System. Int J Mol Sci 2021; 22:ijms22147713. [PMID: 34299332 PMCID: PMC8304141 DOI: 10.3390/ijms22147713] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/08/2021] [Accepted: 07/15/2021] [Indexed: 12/20/2022] Open
Abstract
Exposure of rodents to <20 cGy Space Radiation (SR) impairs performance in several hippocampus-dependent cognitive tasks, including spatial memory. However, there is considerable inter-individual susceptibility to develop SR-induced spatial memory impairment. In this study, a robust label-free mass spectrometry (MS)-based unbiased proteomic profiling approach was used to characterize the composition of the hippocampal proteome in adult male Wistar rats exposed to 15 cGy of 1 GeV/n 48Ti and their sham counterparts. Unique protein signatures were identified in the hippocampal proteome of: (1) sham rats, (2) Ti-exposed rats, (3) Ti-exposed rats that had sham-like spatial memory performance, and (4) Ti-exposed rats that impaired spatial memory performance. Approximately 14% (159) of the proteins detected in hippocampal proteome of sham rats were not detected in the Ti-exposed rats. We explored the possibility that the loss of the Sham-only proteins may arise as a result of SR-induced changes in protein homeostasis. SR-exposure was associated with a switch towards increased pro-ubiquitination proteins from that seen in Sham. These data suggest that the role of the ubiquitin-proteome system as a determinant of SR-induced neurocognitive deficits needs to be more thoroughly investigated.
Collapse
Affiliation(s)
- Alyssa Tidmore
- Department of Radiation Oncology, Eastern Virginia Medical School, 700 W. Olney Rd., Lewis Hall, Norfolk, VA 23507, USA; (A.T.); (A.S.F.); (V.D.D.)
- Department of Microbiology and Molecular Cell Biology; Eastern Virginia Medical School, Norfolk, VA 23507, USA;
- Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA 23507, USA
- Center for Integrative Neuroinflammatory and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Sucharita M. Dutta
- Department of Microbiology and Molecular Cell Biology; Eastern Virginia Medical School, Norfolk, VA 23507, USA;
- Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Arriyam S. Fesshaye
- Department of Radiation Oncology, Eastern Virginia Medical School, 700 W. Olney Rd., Lewis Hall, Norfolk, VA 23507, USA; (A.T.); (A.S.F.); (V.D.D.)
- Department of Microbiology and Molecular Cell Biology; Eastern Virginia Medical School, Norfolk, VA 23507, USA;
- Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA 23507, USA
- Center for Integrative Neuroinflammatory and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - William K. Russell
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA;
| | - Vania D. Duncan
- Department of Radiation Oncology, Eastern Virginia Medical School, 700 W. Olney Rd., Lewis Hall, Norfolk, VA 23507, USA; (A.T.); (A.S.F.); (V.D.D.)
| | - Richard A. Britten
- Department of Radiation Oncology, Eastern Virginia Medical School, 700 W. Olney Rd., Lewis Hall, Norfolk, VA 23507, USA; (A.T.); (A.S.F.); (V.D.D.)
- Department of Microbiology and Molecular Cell Biology; Eastern Virginia Medical School, Norfolk, VA 23507, USA;
- Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA 23507, USA
- Center for Integrative Neuroinflammatory and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA 23507, USA
- Correspondence:
| |
Collapse
|
27
|
Garrett L, Ung MC, Einicke J, Zimprich A, Fenzl F, Pawliczek D, Graw J, Dalke C, Hölter SM. Complex Long-term Effects of Radiation on Adult Mouse Behavior. Radiat Res 2021; 197:67-77. [PMID: 34237145 DOI: 10.1667/rade-20-00281.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 05/24/2021] [Indexed: 11/03/2022]
Abstract
We have shown previously that a single radiation event (0.063, 0.125 or 0.5 Gy, 0.063 Gy/min) in adult mice (age 10 weeks) can have delayed dose-dependent effects on locomotor behavior 18 months postirradiation. The highest dose (0.5 Gy) reduced, whereas the lowest dose (0.063 Gy) increased locomotor activity at older age independent of sex or genotype. In the current study we investigated whether higher doses administered at a higher dose rate (0.5, 1 or 2 Gy, 0.3 Gy/min) at the same age (10 weeks) cause stronger or earlier effects on a range of behaviors, including locomotion, anxiety, sensorimotor and cognitive behavior. There were clear dose-dependent effects on spontaneous locomotor and exploratory activity, anxiety-related behavior, body weight and affiliative social behavior independent of sex or genotype of wild-type and Ercc2S737P heterozygous mice on a mixed C57BL/6JG and C3HeB/FeJ background. In addition, smaller genotype- and dose-dependent radiation effects on working memory were evident in males, but not in females. The strongest dose-dependent radiation effects were present 4 months postirradiation, but only effects on affiliative social behaviors persisted until 12 months postirradiation. The observed radiation-induced behavioral changes were not related to alterations in the eye lens, as 4 months postirradiation anterior and posterior parts of the lens were still normal. Overall, we did not find any sensitizing effect of the mutation towards radiation effects in vivo.
Collapse
Affiliation(s)
- Lillian Garrett
- Helmholtz Zentrum München, German Research Centre for Environmental Health, Institute of Developmental Genetics, Neuherberg, Germany
| | - Marie-Claire Ung
- Helmholtz Zentrum München, German Research Centre for Environmental Health, Institute of Developmental Genetics, Neuherberg, Germany
| | - Jan Einicke
- Helmholtz Zentrum München, German Research Centre for Environmental Health, Institute of Developmental Genetics, Neuherberg, Germany
| | - Annemarie Zimprich
- Technical University Munich, School of Life Science Weihenstephan, Freising, Germany
| | - Felix Fenzl
- Technical University Munich, School of Life Science Weihenstephan, Freising, Germany
| | - Daniel Pawliczek
- Helmholtz Zentrum München, German Research Centre for Environmental Health, Institute of Developmental Genetics, Neuherberg, Germany
| | - Jochen Graw
- Helmholtz Zentrum München, German Research Centre for Environmental Health, Institute of Developmental Genetics, Neuherberg, Germany
| | - Claudia Dalke
- Helmholtz Zentrum München, German Research Centre for Environmental Health, Institute of Developmental Genetics, Neuherberg, Germany
| | - Sabine M Hölter
- Helmholtz Zentrum München, German Research Centre for Environmental Health, Institute of Developmental Genetics, Neuherberg, Germany.,Technical University Munich, School of Life Science Weihenstephan, Freising, Germany
| |
Collapse
|
28
|
Boerma M, Davis CM, Jackson IL, Schaue D, Williams JP. All for one, though not one for all: team players in normal tissue radiobiology. Int J Radiat Biol 2021; 98:346-366. [PMID: 34129427 PMCID: PMC8781287 DOI: 10.1080/09553002.2021.1941383] [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: 03/26/2021] [Revised: 05/27/2021] [Accepted: 05/27/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE As part of the special issue on 'Women in Science', this review offers a perspective on past and ongoing work in the field of normal (non-cancer) tissue radiation biology, highlighting the work of many of the leading contributors to this field of research. We discuss some of the hypotheses that have guided investigations, with a focus on some of the critical organs considered dose-limiting with respect to radiation therapy, and speculate on where the field needs to go in the future. CONCLUSIONS The scope of work that makes up normal tissue radiation biology has and continues to play a pivotal role in the radiation sciences, ensuring the most effective application of radiation in imaging and therapy, as well as contributing to radiation protection efforts. However, despite the proven historical value of preclinical findings, recent decades have seen clinical practice move ahead with altered fractionation scheduling based on empirical observations, with little to no (or even negative) supporting scientific data. Given our current appreciation of the complexity of normal tissue radiation responses and their temporal variability, with tissue- and/or organ-specific mechanisms that include intra-, inter- and extracellular messaging, as well as contributions from systemic compartments, such as the immune system, the need to maintain a positive therapeutic ratio has never been more urgent. Importantly, mitigation and treatment strategies, whether for the clinic, emergency use following accidental or deliberate releases, or reducing occupational risk, will likely require multi-targeted approaches that involve both local and systemic intervention. From our personal perspective as five 'Women in Science', we would like to acknowledge and applaud the role that many female scientists have played in this field. We stand on the shoulders of those who have gone before, some of whom are fellow contributors to this special issue.
Collapse
Affiliation(s)
- Marjan Boerma
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Catherine M. Davis
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Isabel L. Jackson
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Dörthe Schaue
- Department of Radiation Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | - Jacqueline P. Williams
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
29
|
Martinez NE, Jokisch DW, Dauer LT, Eckerman KF, Goans RE, Brockman JD, Tolmachev SY, Avtandilashvili M, Mumma MT, Boice JD, Leggett RW. Radium dial workers: back to the future. Int J Radiat Biol 2021; 98:750-768. [PMID: 33900890 PMCID: PMC10563809 DOI: 10.1080/09553002.2021.1917785] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 04/01/2021] [Accepted: 04/02/2021] [Indexed: 12/16/2022]
Abstract
PURPOSE This paper reviews the history of the radium dial workers in the United States, summarizes the scientific progress made since the last evaluation in the early 1990s, and discusses current progress in updating the epidemiologic cohort and applying new dosimetric models for radiation risk assessment. BACKGROUND The discoveries of radiation and radioactivity led quickly to medical and commercial applications at the turn of the 20th century, including the development of radioluminescent paint, made by combining radium with phosphorescent material and adhesive. Workers involved with the painting of dials and instruments included painters, handlers, ancillary workers, and chemists who fabricated the paint. Dial painters were primarily women and, prior to the mid to late 1920s, would use their lips to give the brush a fine point, resulting in high intakes of radium. The tragic experience of the dial painters had a significant impact on industrial safety standards, including protection measures taken during the Manhattan Project. The dial workers study has formed the basis for radiation protection standards for intakes of radionuclides by workers and the public. EPIDEMIOLOGIC APPROACH The mortality experience of 3,276 radium dial painters and handlers employed between 1913 and 1949 is being determined through 2019. The last epidemiologic follow-up was 30 years ago when most of these workers were still alive. Nearly 65% were born before 1920, 37.5% were teenagers when first hired, and nearly 50% were hired before 1930 when the habit of placing brushes in mouths essentially stopped. Comprehensive dose reconstruction techniques are being applied to estimate organ doses for each worker related to the intake of 226Ra, 228Ra, and associated photon exposures. Time dependent dose-response analyses will estimate lifetime risks for specific causes of death. DISCUSSION The study of radium dial workers is part of the Million Person Study of low-dose health effects that is designed to evaluate radiation risks among healthy American workers and veterans. Despite being one of the most important and influential radiation effects studies ever conducted, shifting programmatic responsibilities and declining funding led to the termination of the radium program of studies in the early 1990s. Renewed interest and opportunity have arisen. With scientific progress made in dosimetric methodology and models, the ability to perform a study over the entire life span, and the potential applicability to other scenarios such as medicine, environmental contamination and space exploration, the radium dial workers have once again come to the forefront.
Collapse
Affiliation(s)
- Nicole E. Martinez
- Department of Environmental Engineering and Earth Sciences, Clemson University, Clemson, SC, USA
- Center for Radiation Protection Knowledge, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - Derek W. Jokisch
- Center for Radiation Protection Knowledge, Oak Ridge National Laboratory, Oak Ridge, TN, USA
- Department of Physics and Engineering, Francis Marion University, Florence, SC, USA
| | - Lawrence T. Dauer
- Department of Medical Physics and Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Keith F. Eckerman
- Center for Radiation Protection Knowledge, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | | | - John D. Brockman
- Department of Chemistry, University of Missouri, Columbia, MO, USA
| | - Sergey Y. Tolmachev
- United States Transuranium and Uranium Registries, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Richland, WA, USA
| | - Maia Avtandilashvili
- United States Transuranium and Uranium Registries, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Richland, WA, USA
| | - Michael T. Mumma
- International Epidemiology Institute, Rockville, MD, USA
- Department of Medicine, Division of Epidemiology, Vanderbilt Epidemiology Center and Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | - John D. Boice
- Department of Medicine, Division of Epidemiology, Vanderbilt Epidemiology Center and Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
- National Council on Radiation Protection and Measurements, Bethesda, MD, USA
| | - Richard W. Leggett
- Center for Radiation Protection Knowledge, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| |
Collapse
|
30
|
Britten RA, Wellman LL, Sanford LD. Progressive increase in the complexity and translatability of rodent testing to assess space-radiation induced cognitive impairment. Neurosci Biobehav Rev 2021; 126:159-174. [PMID: 33766676 DOI: 10.1016/j.neubiorev.2021.01.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 12/15/2020] [Accepted: 01/07/2021] [Indexed: 11/29/2022]
Abstract
Ground-based rodent models have established that space radiation doses (approximately those that astronauts will be exposed to on a mission to Mars) significantly impair performance in a wide range of cognitive tasks. Over the last 40 years there has been a progressive increase in both the complexity and the translatability (to humans) of the cognitive tasks investigated. This review outlines technical and conceptual advances in space radiation rodent testing approaches, along with the advances in analytical approaches, that will make data from ground based studies more amenable to probabilistic risk analysis. While great progress has been made in determining the impact of space radiation on many advanced cognitive processes, challenges remain that need to be addressed prior to commencing deep space missions. A summary of on-going attempts to address existing knowledge gaps and the critical role that rodent studies will have in establishing the impact of space radiation on even more complex (human) cognitive tasks are presented and discussed.
Collapse
Affiliation(s)
- Richard A Britten
- Department of Radiation Oncology, Eastern Virginia Medical School, Norfolk, VA, 23507, USA; Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, 23507, USA; Leroy T Canoles Jr. Cancer Center, Eastern Virginia Medical School, Norfolk, VA, 23507, USA; Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA, 23507, USA.
| | - Laurie L Wellman
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA, 23507, USA; Department of Pathology & Anatomy, Eastern Virginia Medical School, Norfolk, VA, 23507, USA
| | - Larry D Sanford
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA, 23507, USA; Department of Pathology & Anatomy, Eastern Virginia Medical School, Norfolk, VA, 23507, USA
| |
Collapse
|