51
|
Kiffer F, Howe AK, Carr H, Wang J, Alexander T, Anderson JE, Groves T, Seawright JW, Sridharan V, Carter G, Boerma M, Allen AR. Late effects of 1H irradiation on hippocampal physiology. LIFE SCIENCES IN SPACE RESEARCH 2018; 17:51-62. [PMID: 29753414 PMCID: PMC7063743 DOI: 10.1016/j.lssr.2018.03.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 03/07/2018] [Accepted: 03/10/2018] [Indexed: 05/21/2023]
Abstract
NASA's Missions to Mars and beyond will expose flight crews to potentially dangerous levels of charged-particle radiation. Of all charged nuclei, 1H is the most abundant charged particle in both the galactic cosmic ray (GCR) and solar particle event (SPE) spectra. There are currently no functional spacecraft shielding materials that are able to mitigate the charged-particle radiation encountered in space. Recent studies have demonstrated cognitive injuries due to high-dose 1H exposures in rodents. Our study investigated the effects of 1H irradiation on neuronal morphology in the hippocampus of adult male mice. 6-month-old mice received whole-body exposure to 1H at 0.5 and 1 Gy (150 MeV/n; 0.35-0.55 Gy/min) at NASA's Space Radiation Laboratory in Upton, NY. At 9-months post-irradiation, we tested each animal's open-field exploratory performance. After sacrifice, we dissected the brains along the midsagittal plane, and then either fixed or dissected further and snap-froze them. Our data showed that exposure to 0.5 Gy or 1 Gy 1H significantly increased animals' anxiety behavior in open-field testing. Our micromorphometric analyses revealed significant decreases in mushroom spine density and dendrite morphology in the Dentate Gyrus, Cornu Ammonis 3 and 1 of the hippocampus, and lowered expression of synaptic markers. Our data suggest 1H radiation significantly increased exploration anxiety and modulated the dendritic spine and dendrite morphology of hippocampal neurons at a dose of 0.5 or 1 Gy.
Collapse
Affiliation(s)
- Frederico Kiffer
- Division of Radiation Health at the University of Arkansas for Medical Sciences, 4301 West Markham, Suite 441B-2, Little Rock, AR 72205, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States.
| | - Alexis K Howe
- Division of Radiation Health at the University of Arkansas for Medical Sciences, 4301 West Markham, Suite 441B-2, Little Rock, AR 72205, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States.
| | - Hannah Carr
- Division of Radiation Health at the University of Arkansas for Medical Sciences, 4301 West Markham, Suite 441B-2, Little Rock, AR 72205, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States.
| | - Jing Wang
- Division of Radiation Health at the University of Arkansas for Medical Sciences, 4301 West Markham, Suite 441B-2, Little Rock, AR 72205, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States.
| | - Tyler Alexander
- Division of Radiation Health at the University of Arkansas for Medical Sciences, 4301 West Markham, Suite 441B-2, Little Rock, AR 72205, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States.
| | - Julie E Anderson
- Division of Radiation Health at the University of Arkansas for Medical Sciences, 4301 West Markham, Suite 441B-2, Little Rock, AR 72205, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States.
| | - Thomas Groves
- Division of Radiation Health at the University of Arkansas for Medical Sciences, 4301 West Markham, Suite 441B-2, Little Rock, AR 72205, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States; Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States.
| | - John W Seawright
- Division of Radiation Health at the University of Arkansas for Medical Sciences, 4301 West Markham, Suite 441B-2, Little Rock, AR 72205, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States.
| | - Vijayalakshmi Sridharan
- Division of Radiation Health at the University of Arkansas for Medical Sciences, 4301 West Markham, Suite 441B-2, Little Rock, AR 72205, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States.
| | - Gwendolyn Carter
- Division of Radiation Health at the University of Arkansas for Medical Sciences, 4301 West Markham, Suite 441B-2, Little Rock, AR 72205, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States.
| | - Marjan Boerma
- Division of Radiation Health at the University of Arkansas for Medical Sciences, 4301 West Markham, Suite 441B-2, Little Rock, AR 72205, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States.
| | - Antiño R Allen
- Division of Radiation Health at the University of Arkansas for Medical Sciences, 4301 West Markham, Suite 441B-2, Little Rock, AR 72205, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States; Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States.
| |
Collapse
|
52
|
Cacao E, Parihar VK, Limoli CL, Cucinotta FA. Stochastic Modeling of Radiation-induced Dendritic Damage on in silico Mouse Hippocampal Neurons. Sci Rep 2018; 8:5494. [PMID: 29615729 PMCID: PMC5882641 DOI: 10.1038/s41598-018-23855-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 03/21/2018] [Indexed: 12/20/2022] Open
Abstract
Cognitive dysfunction associated with radiotherapy for cancer treatment has been correlated to several factors, one of which is changes to the dendritic morphology of neuronal cells. Alterations in dendritic geometry and branching patterns are often accompanied by deficits that impact learning and memory. The purpose of this study is to develop a novel predictive model of neuronal dendritic damages caused by exposure to low linear energy transfer (LET) radiation, such as X-rays, γ-rays and high-energy protons. We established in silico representations of mouse hippocampal dentate granule cell layer (GCL) and CA1 pyramidal neurons, which are frequently examined in radiation-induced cognitive decrements. The in silico representations are used in a stochastic model that describes time dependent dendritic damage induced by exposure to low LET radiation. Changes in morphometric parameters, such as total dendritic length, number of branch points and branch number, including the Sholl analysis for single neurons are described by the model. Our model based predictions for different patterns of morphological changes based on energy deposition in dendritic segments (EDDS) will serve as a useful basis to compare specific patterns of morphological alterations caused by EDDS mechanisms.
Collapse
Affiliation(s)
- Eliedonna Cacao
- Department of Health Physics and Diagnostic Sciences, University of Nevada, Las Vegas, NV, United States of America
| | - Vipan K Parihar
- Department of Radiation Oncology, University of California, Irvine, CA, United States of America
| | - Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine, CA, United States of America
| | - Francis A Cucinotta
- Department of Health Physics and Diagnostic Sciences, University of Nevada, Las Vegas, NV, United States of America.
| |
Collapse
|
53
|
Allen BD, Acharya MM, Lu C, Giedzinski E, Chmielewski NN, Quach D, Hefferan M, Johe KK, Limoli CL. Remediation of Radiation-Induced Cognitive Dysfunction through Oral Administration of the Neuroprotective Compound NSI-189. Radiat Res 2018; 189:345-353. [PMID: 29351056 DOI: 10.1667/rr14879.1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Clinical management of primary and secondary central nervous system (CNS) malignancies frequently includes radiotherapy to forestall tumor growth and recurrence after surgical resection. While cranial radiotherapy remains beneficial, adult and pediatric brain tumor survivors suffer from a wide range of debilitating and progressive cognitive deficits. Although this has been recognized as a significant problem for decades, there remains no clinical recourse for the unintended neurocognitive sequelae associated with these types of cancer treatments. In previous work, multiple mechanisms have been identified that contribute to radiation-induced cognitive dysfunction, including the inhibition of neurogenesis caused by the depletion of radiosensitive populations of stem and progenitor cells in the hippocampus. To explore the potential neuroprotective properties of a pro-neurogenic compound NSI-189, Long-Evans rats were subjected to a clinically relevant fractionated irradiation protocol followed by four weeks of NSI-189 administered daily by oral gavage. Animals were then subjected to five different behavioral tasks followed by an analysis of neurogenesis, hippocampal volume and neuroinflammation. Irradiated cohorts manifested significant behavioral decrements on all four spontaneous exploration tasks. Importantly, NSI-189 treatment resulted in significantly improved performance in four of these tasks: novel place recognition, novel object recognition, object in place and temporal order. In addition, there was a trend of improved performance in the contextual phase of the fear conditioning task. Importantly, enhanced cognition in the NSI-189-treated cohort was found to persist one month after the cessation of drug treatment. These neurocognitive benefits of NSI-189 coincided with a significant increase in neurogenesis and a significant decrease in the numbers of activated microglia compared to the irradiated cohort that was given vehicle alone. The foregoing changes were not accompanied by major changes in hippocampal volume. These data demonstrate that oral administration of a pro-neurogenic compound exhibiting anti-inflammatory indications could impart long-term neurocognitive benefits in the irradiated brain.
Collapse
Affiliation(s)
- Barrett D Allen
- a Department of Radiation Oncology, University of California, Irvine, Irvine, California 92697-2695
| | - Munjal M Acharya
- a Department of Radiation Oncology, University of California, Irvine, Irvine, California 92697-2695
| | - Celine Lu
- a Department of Radiation Oncology, University of California, Irvine, Irvine, California 92697-2695
| | - Erich Giedzinski
- a Department of Radiation Oncology, University of California, Irvine, Irvine, California 92697-2695
| | - Nicole N Chmielewski
- a Department of Radiation Oncology, University of California, Irvine, Irvine, California 92697-2695
| | - David Quach
- b Neuralstem, Inc., Germantown, Maryland 20876
| | | | - Karl K Johe
- b Neuralstem, Inc., Germantown, Maryland 20876
| | - Charles L Limoli
- a Department of Radiation Oncology, University of California, Irvine, Irvine, California 92697-2695
| |
Collapse
|
54
|
Programming of Cell Resistance to Genotoxic and Oxidative Stress. Biomedicines 2018; 6:biomedicines6010005. [PMID: 29301323 PMCID: PMC5874662 DOI: 10.3390/biomedicines6010005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Revised: 12/23/2017] [Accepted: 12/31/2017] [Indexed: 12/23/2022] Open
Abstract
Different organisms, cell types, and even similar cell lines can dramatically differ in resistance to genotoxic stress. This testifies to the wide opportunities for genetic and epigenetic regulation of stress resistance. These opportunities could be used to increase the effectiveness of cancer therapy, develop new varieties of plants and animals, and search for new pharmacological targets to enhance human radioresistance, which can be used for manned deep space expeditions. Based on the comparison of transcriptomic studies in cancer cells, in this review, we propose that there is a high diversity of genetic mechanisms of development of genotoxic stress resistance. This review focused on possibilities and limitations of the regulation of the resistance of normal cells and whole organisms to genotoxic and oxidative stress by the overexpressing of stress-response genes. Moreover, the existing experimental data on the effect of such overexpression on the resistance of cells and organisms to various genotoxic agents has been analyzed and systematized. We suggest that the recent advances in the development of multiplex and highly customizable gene overexpression technology that utilizes the mutant Cas9 protein and the abundance of available data on gene functions and their signal networks open new opportunities for research in this field.
Collapse
|
55
|
Liao X, Huang C, Zhang D, Wang J, Li J, Jin H, Huang C. Mitochondrial catalase induces cells transformation through nucleolin-dependent Cox-2 mRNA stabilization. Free Radic Biol Med 2017; 113:478-486. [PMID: 29097213 DOI: 10.1016/j.freeradbiomed.2017.10.387] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Revised: 10/26/2017] [Accepted: 10/27/2017] [Indexed: 12/12/2022]
Abstract
It's well documented that over-production of reactive oxygen species (ROS) causes detrimental damages to cells. While a low level of ROS, such as H2O2, functions as signaling transducer and motivates cell proliferation in both cancer and non-transformed stem cells. As a double-edged sword, the direct evidence for demonstrating the function of H2O2 in the cause of tumor is barely characterized in intact cells. In our current study, we found that targeted expression of mitochondrial catalase (mCAT), but not catalase, could significantly reduce the accumulation of H2O2 in mouse epithelial JB6 Cl41 cells, consequently led to the cell malignant transformation and anchorage-independent cell growth. Further study revealed that this reduction of H2O2 resulted in the translocation of nucleolin from the cytoplasm to nuclear, and maintaining the nucleolin nuclear location status, and in turn stabilizing the cox-2 mRNA and consequently leading to a COX-2 protein upregulation, as well as malignant transforming mCAT-overexpressed Cl41 cells. Collectively, our studies here provide direct experimental evidence demonstrating a novel function and molecular mechanisms of mCAT in transforming mouse Cl41 cells, and high significance insight into understanding the beneficial aspect of H2O2 in circumventing tumor promotion and the theoretical basis for the management of H2O2 in the clinic implementation as a chemotherapeutic strategy.
Collapse
Affiliation(s)
- Xin Liao
- Nelson Institute of Environmental Medicine, New York University, School of Medicine, Tuxedo Park, NY 10987, USA; Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chao Huang
- Nelson Institute of Environmental Medicine, New York University, School of Medicine, Tuxedo Park, NY 10987, USA
| | - Dongyun Zhang
- Nelson Institute of Environmental Medicine, New York University, School of Medicine, Tuxedo Park, NY 10987, USA
| | - Jingjing Wang
- Nelson Institute of Environmental Medicine, New York University, School of Medicine, Tuxedo Park, NY 10987, USA; Zhejiang Provincial Key Laboratory for Technology & Application of Model Organisms, School of Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jingxia Li
- Nelson Institute of Environmental Medicine, New York University, School of Medicine, Tuxedo Park, NY 10987, USA
| | - Honglei Jin
- Zhejiang Provincial Key Laboratory for Technology & Application of Model Organisms, School of Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Chuanshu Huang
- Nelson Institute of Environmental Medicine, New York University, School of Medicine, Tuxedo Park, NY 10987, USA.
| |
Collapse
|
56
|
Rudobeck E, Bellone JA, Szücs A, Bonnick K, Mehrotra-Carter S, Badaut J, Nelson GA, Hartman RE, Vlkolinský R. Low-dose proton radiation effects in a transgenic mouse model of Alzheimer's disease - Implications for space travel. PLoS One 2017; 12:e0186168. [PMID: 29186131 PMCID: PMC5706673 DOI: 10.1371/journal.pone.0186168] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 09/26/2017] [Indexed: 12/13/2022] Open
Abstract
Space radiation represents a significant health risk for astronauts. Ground-based animal studies indicate that space radiation affects neuronal functions such as excitability, synaptic transmission, and plasticity, and it may accelerate the onset of Alzheimer's disease (AD). Although protons represent the main constituent in the space radiation spectrum, their effects on AD-related pathology have not been tested. We irradiated 3 month-old APP/PSEN1 transgenic (TG) and wild type (WT) mice with protons (150 MeV; 0.1-1.0 Gy; whole body) and evaluated functional and biochemical hallmarks of AD. We performed behavioral tests in the water maze (WM) before irradiation and in the WM and Barnes maze at 3 and 6 months post-irradiation to evaluate spatial learning and memory. We also performed electrophysiological recordings in vitro in hippocampal slices prepared 6 and 9 months post-irradiation to evaluate excitatory synaptic transmission and plasticity. Next, we evaluated amyloid β (Aβ) deposition in the contralateral hippocampus and adjacent cortex using immunohistochemistry. In cortical homogenates, we analyzed the levels of the presynaptic marker synaptophysin by Western blotting and measured pro-inflammatory cytokine levels (TNFα, IL-1β, IL-6, CXCL10 and CCL2) by bead-based multiplex assay. TG mice performed significantly worse than WT mice in the WM. Irradiation of TG mice did not affect their behavioral performance, but reduced the amplitudes of population spikes and inhibited paired-pulse facilitation in CA1 neurons. These electrophysiological alterations in the TG mice were qualitatively different from those observed in WT mice, in which irradiation increased excitability and synaptic efficacy. Irradiation increased Aβ deposition in the cortex of TG mice without affecting cytokine levels and increased synaptophysin expression in WT mice (but not in the TG mice). Although irradiation with protons increased Aβ deposition, the complex functional and biochemical results indicate that irradiation effects are not synergistic to AD pathology.
Collapse
Affiliation(s)
- Emil Rudobeck
- Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, United States of America
| | - John A. Bellone
- Department of Psychology, School of Behavioral Health, Loma Linda University, Loma Linda, CA, United States of America
| | - Attila Szücs
- BioCircuits Institute, University of California San Diego, La Jolla, CA, United States of America
| | - Kristine Bonnick
- Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, United States of America
| | - Shalini Mehrotra-Carter
- Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, United States of America
| | - Jerome Badaut
- Department of Physiology, School of Medicine, Loma Linda University, Loma Linda, CA, United States of America
| | - Gregory A. Nelson
- Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, United States of America
| | - Richard E. Hartman
- Department of Psychology, School of Behavioral Health, Loma Linda University, Loma Linda, CA, United States of America
| | - Roman Vlkolinský
- Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, United States of America
| |
Collapse
|
57
|
Whoolery CW, Walker AK, Richardson DR, Lucero MJ, Reynolds RP, Beddow DH, Clark KL, Shih HY, LeBlanc JA, Cole MG, Amaral WZ, Mukherjee S, Zhang S, Ahn F, Bulin SE, DeCarolis NA, Rivera PD, Chen BPC, Yun S, Eisch AJ. Whole-Body Exposure to 28Si-Radiation Dose-Dependently Disrupts Dentate Gyrus Neurogenesis and Proliferation in the Short Term and New Neuron Survival and Contextual Fear Conditioning in the Long Term. Radiat Res 2017; 188:532-551. [PMID: 28945526 PMCID: PMC5901735 DOI: 10.1667/rr14797.1] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Astronauts traveling to Mars will be exposed to chronic low doses of galactic cosmic space radiation, which contains highly charged, high-energy (HZE) particles. 56Fe-HZE-particle exposure decreases hippocampal dentate gyrus (DG) neurogenesis and disrupts hippocampal function in young adult rodents, raising the possibility of impaired astronaut cognition and risk of mission failure. However, far less is known about how exposure to other HZE particles, such as 28Si, influences hippocampal neurogenesis and function. To compare the influence of 28Si exposure on indices of neurogenesis and hippocampal function with previous studies on 56Fe exposure, 9-week-old C57BL/6J and Nestin-GFP mice (NGFP; made and maintained for 10 or more generations on a C57BL/6J background) received whole-body 28Si-particle-radiation exposure (0, 0.2 and 1 Gy, 300 MeV/n, LET 67 KeV/μ, dose rate 1 Gy/min). For neurogenesis assessment, the NGFP mice were injected with the mitotic marker BrdU at 22 h postirradiation and brains were examined for indices of hippocampal proliferation and neurogenesis, including Ki67+, BrdU+, BrdU+NeuN+ and DCX+ cell numbers at short- and long-term time points (24 h and 3 months postirradiation, respectively). In the short-term group, stereology revealed fewer Ki67+, BrdU+ and DCX+ cells in 1-Gy-irradiated group relative to nonirradiated control mice, fewer Ki67+ and DCX+ cells in 0.2 Gy group relative to control group and fewer BrdU+ and DCX+ cells in 1 Gy group relative to 0.2 Gy group. In contrast to the clearly observed radiation-induced, dose-dependent reductions in the short-term group across all markers, only a few neurogenesis indices were changed in the long-term irradiated groups. Notably, there were fewer surviving BrdU+ cells in the 1 Gy group relative to 0- and 0.2-Gy-irradiated mice in the long-term group. When the short- and long-term groups were analyzed by sex, exposure to radiation had a similar effect on neurogenesis indices in male and female mice, although only male mice showed fewer surviving BrdU+ cells in the long-term group. Fluorescent immunolabeling and confocal phenotypic analysis revealed that most surviving BrdU+ cells in the long-term group expressed the neuronal marker NeuN, definitively confirming that exposure to 1 Gy 28Si radiation decreased the number of surviving adult-generated neurons in male mice relative to both 0- and 0.2-Gy-irradiated mice. For hippocampal function assessment, 9-week-old male C57BL/6J mice received whole-body 28Si-particle exposure and were then assessed long-term for performance on contextual and cued fear conditioning. In the context test the animals that received 0.2 Gy froze less relative to control animals, suggesting decreased hippocampal-dependent function. However, in the cued fear conditioning test, animals that received 1 Gy froze more during the pretone portion of the test, relative to controls and 0.2-Gy-irradiated mice, suggesting enhanced anxiety. Compared to previously reported studies, these data suggest that 28Si-radiation exposure damages neurogenesis, but to a lesser extent than 56Fe radiation and that low-dose 28Si exposure induces abnormalities in hippocampal function, disrupting fear memory but also inducing anxiety-like behavior. Furthermore, exposure to 28Si radiation decreased new neuron survival in long-term male groups but not females suggests that sex may be an important factor when performing brain health risk assessment for astronauts traveling in space.
Collapse
Affiliation(s)
- Cody W. Whoolery
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, Texas
| | - Angela K. Walker
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, Texas
| | | | - Melanie J. Lucero
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, Texas
| | - Ryan P. Reynolds
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, Texas
- Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - David H. Beddow
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, Texas
| | - K. Lyles Clark
- Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Hung-Ying Shih
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, Texas
| | - Junie A. LeBlanc
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, Texas
| | - Mara G. Cole
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, Texas
| | | | - Shibani Mukherjee
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, Texas
| | - Shichuan Zhang
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, Texas
| | - Francisca Ahn
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, Texas
| | - Sarah E. Bulin
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, Texas
| | | | - Phillip D. Rivera
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, Texas
| | - Benjamin P. C. Chen
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, Texas
| | - Sanghee Yun
- Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Amelia J. Eisch
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, Texas
- Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| |
Collapse
|
58
|
Tahimic CGT, Globus RK. Redox Signaling and Its Impact on Skeletal and Vascular Responses to Spaceflight. Int J Mol Sci 2017; 18:ijms18102153. [PMID: 29035346 PMCID: PMC5666834 DOI: 10.3390/ijms18102153] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Revised: 09/30/2017] [Accepted: 10/10/2017] [Indexed: 12/16/2022] Open
Abstract
Spaceflight entails exposure to numerous environmental challenges with the potential to contribute to both musculoskeletal and vascular dysfunction. The purpose of this review is to describe current understanding of microgravity and radiation impacts on the mammalian skeleton and associated vasculature at the level of the whole organism. Recent experiments from spaceflight and ground-based models have provided fresh insights into how these environmental stresses influence mechanisms that are related to redox signaling, oxidative stress, and tissue dysfunction. Emerging mechanistic knowledge on cellular defenses to radiation and other environmental stressors, including microgravity, are useful for both screening and developing interventions against spaceflight-induced deficits in bone and vascular function.
Collapse
Affiliation(s)
- Candice G T Tahimic
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA.
- KBRWyle, Moffett Field, CA 94035, USA.
| | - Ruth K Globus
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA.
| |
Collapse
|
59
|
Abstract
Purpose of review To encapsulate past and current research efforts focused on stem cell transplantation strategies to resolve radiation-induced cognitive dysfunction. Recent Findings Transplantation of human stem cells in the irradiated brain was first shown to resolve radiation-induced cognitive dysfunction in a landmark paper by Acharya et al., appearing in PNAS in 2009. Since that time, work from the same laboratory as well as other groups have reported on the beneficial (as well as detrimental) effects of stem cell grafting after cranial radiation exposure. Improved learning and memory found many months after engraftment has since been associated with a preservation of host neuronal morphology, a suppression of neuroinflammation, improved myelination and increased cerebral blood flow. Interestingly, many (if not all) of these beneficial effects can be demonstrated by substituting stem cells with microvesicles derived from human stem cells during transplantation, thereby eliminating many of the more long-standing concerns related to immunorejection and teratoma formation. Summary Stem cell and microvesicle transplantation into the irradiated brain of rodents has uncovered some unexpected benefits that hold promise for ameliorating many of adverse neurocognitive complications associated with major cancer treatments. Properly developed, such approaches may provide much needed clinical recourse to millions of cancer survivors suffering from the unintended side effects of their cancer therapies.
Collapse
|
60
|
Decrock E, Hoorelbeke D, Ramadan R, Delvaeye T, De Bock M, Wang N, Krysko DV, Baatout S, Bultynck G, Aerts A, Vinken M, Leybaert L. Calcium, oxidative stress and connexin channels, a harmonious orchestra directing the response to radiotherapy treatment? BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:1099-1120. [DOI: 10.1016/j.bbamcr.2017.02.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 02/02/2017] [Accepted: 02/04/2017] [Indexed: 02/07/2023]
|
61
|
CNS bioavailability and radiation protection of normal hippocampal neurogenesis by a lipophilic Mn porphyrin-based superoxide dismutase mimic, MnTnBuOE-2-PyP 5. Redox Biol 2017; 12:864-871. [PMID: 28454069 PMCID: PMC5407575 DOI: 10.1016/j.redox.2017.04.027] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Revised: 04/12/2017] [Accepted: 04/13/2017] [Indexed: 12/25/2022] Open
Abstract
Although radiation therapy can be effective against cancer, potential damage to normal tissues limits the amount that can be safely administered. In central nervous system (CNS), radiation damage to normal tissues is presented, in part, as suppressed hippocampal neurogenesis and impaired cognitive functions. Mn porphyrin (MnP)-based redox active drugs have demonstrated differential effects on cancer and normal tissues in experimental animals that lead to protection of normal tissues and radio- and chemo-sensitization of cancers. To test the efficacy of MnPs in CNS radioprotection, we first examined the tissue levels of three different MnPs – MnTE-2-PyP5+(MnE), MnTnHex-2-PyP5+(MnHex), and MnTnBuOE-2-PyP5+(MnBuOE). Nanomolar concentrations of MnHex and MnBuOE were detected in various brain regions after daily subcutaneous administration, and MnBuOE was well tolerated at a daily dose of 3 mg/kg. Administration of MnBuOE for one week before cranial irradiation and continued for one week afterwards supported production and long-term survival of newborn neurons in the hippocampal dentate gyrus. MnP-driven S-glutathionylation in cortex and hippocampus showed differential responses to MnP administration and radiation in these two brain regions. A better understanding of how preserved hippocampal neurogenesis correlates with cognitive functions following cranial irradiation will be helpful in designing better MnP-based radioprotection strategies. Bioavailability of MnPs in individual brain regions were determined by LC-MS/MS. CNS MnBuOE and MnHex levels were between 15 and 160 nM after daily administration. MnBuOE administration ameliorated radiation effects on hippocampal neurogenesis. MnBuOE preserved categories E&F Dcx+ neurons after cranial irradiation. MnBuOE and irradiation lead to changes in protein S-glutathionylation in the CNS.
Collapse
|
62
|
Mitochondrial-Targeted Catalase: Extended Longevity and the Roles in Various Disease Models. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 146:203-241. [PMID: 28253986 DOI: 10.1016/bs.pmbts.2016.12.015] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The free-radical theory of aging was proposed more than 50 years ago. As one of the most popular mechanisms explaining the aging process, it has been extensively studied in several model organisms. However, the results remain controversial. The mitochondrial version of free-radical theory of aging proposes that mitochondria are both the primary sources of reactive oxygen species (ROS) and the primary targets of ROS-induced damage. One critical ROS is hydrogen peroxide, which is naturally degraded by catalase in peroxisomes or glutathione peroxidase within mitochondria. Our laboratory developed mice-overexpressing catalase targeted to mitochondria (mCAT), peroxisomes (pCAT), or the nucleus (nCAT) in order to investigate the role of hydrogen peroxide in different subcellular compartments in aging and age-related diseases. The mCAT mice have demonstrated the largest effects on life span and healthspan extension. This chapter will discuss the mCAT phenotype and review studies using mCAT to investigate the roles of mitochondrial oxidative stresses in various disease models, including metabolic syndrome and atherosclerosis, cardiac aging, heart failure, skeletal muscle pathology, sensory defect, neurodegenerative diseases, and cancer. As ROS has been increasingly recognized as essential signaling molecules that may be beneficial in hormesis, stress response and immunity, the potential pleiotropic, or adverse effects of mCAT are also discussed. Finally, the development of small-molecule mitochondrial-targeted therapeutic approaches is reviewed.
Collapse
|
63
|
Alwood JS, Ronca AE, Mains RC, Shelhamer MJ, Smith JD, Goodwin TJ. From the bench to exploration medicine: NASA life sciences translational research for human exploration and habitation missions. NPJ Microgravity 2017. [PMID: 28649627 PMCID: PMC5460236 DOI: 10.1038/s41526-016-0002-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
NASA’s Space Biology and Human Research Program entities have recently spearheaded communications both internally and externally to coordinate the agency’s translational research efforts. In this paper, we strongly advocate for translational research at NASA, provide recent examples of NASA sponsored early-stage translational research, and discuss options for a path forward. Our overall objective is to help in stimulating a collaborative research across multiple disciplines and entities that, working together, will more effectively and more rapidly achieve NASA’s goals for human spaceflight.
Collapse
Affiliation(s)
- Joshua S Alwood
- Space BioSciences Division, NASA Ames Research Center, Moffett Field, CA USA
| | - April E Ronca
- Space BioSciences Division, NASA Ames Research Center, Moffett Field, CA USA.,Wake Forest School of Medicine, Winston-Salem, NC USA
| | | | - Mark J Shelhamer
- Human Research Program, NASA Johnson Space Center, Houston, TX USA
| | - Jeffrey D Smith
- Space BioSciences Division, NASA Ames Research Center, Moffett Field, CA USA
| | - Thomas J Goodwin
- Biomedical Research and Environmental Sciences Division, NASA Johnson Space Center, Houston, TX USA
| |
Collapse
|
64
|
Kim SJ, Cheresh P, Jablonski RP, Morales-Nebreda L, Cheng Y, Hogan E, Yeldandi A, Chi M, Piseaux R, Ridge K, Michael Hart C, Chandel N, Scott Budinger GR, Kamp DW. Mitochondrial catalase overexpressed transgenic mice are protected against lung fibrosis in part via preventing alveolar epithelial cell mitochondrial DNA damage. Free Radic Biol Med 2016; 101:482-490. [PMID: 27840320 PMCID: PMC5928521 DOI: 10.1016/j.freeradbiomed.2016.11.007] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 10/31/2016] [Accepted: 11/01/2016] [Indexed: 12/27/2022]
Abstract
RATIONALE Alveolar epithelial cell (AEC) injury and mitochondrial dysfunction are important in the development of lung fibrosis. Our group has shown that in the asbestos exposed lung, the generation of mitochondrial reactive oxygen species (ROS) in AEC mediate mitochondrial DNA (mtDNA) damage and apoptosis which are necessary for lung fibrosis. These data suggest that mitochondrial-targeted antioxidants should ameliorate asbestos-induced lung. OBJECTIVE To determine whether transgenic mice that express mitochondrial-targeted catalase (MCAT) have reduced lung fibrosis following exposure to asbestos or bleomycin and, if so, whether this occurs in association with reduced AEC mtDNA damage and apoptosis. METHODS Crocidolite asbestos (100µg/50µL), TiO2 (negative control), bleomycin (0.025 units/50µL), or PBS was instilled intratracheally in 8-10 week-old wild-type (WT - C57Bl/6J) or MCAT mice. The lungs were harvested at 21d. Lung fibrosis was quantified by collagen levels (Sircol) and lung fibrosis scores. AEC apoptosis was assessed by cleaved caspase-3 (CC-3)/Surfactant protein C (SFTPC) immunohistochemistry (IHC) and semi-quantitative analysis. AEC (primary AT2 cells from WT and MCAT mice and MLE-12 cells) mtDNA damage was assessed by a quantitative PCR-based assay, apoptosis was assessed by DNA fragmentation, and ROS production was assessed by a Mito-Sox assay. RESULTS Compared to WT, crocidolite-exposed MCAT mice exhibit reduced pulmonary fibrosis as measured by lung collagen levels and lung fibrosis score. The protective effects in MCAT mice were accompanied by reduced AEC mtDNA damage and apoptosis. Similar findings were noted following bleomycin exposure. Euk-134, a mitochondrial SOD/catalase mimetic, attenuated MLE-12 cell DNA damage and apoptosis. Finally, compared to WT, asbestos-induced MCAT AT2 cell ROS production was reduced. CONCLUSIONS Our finding that MCAT mice have reduced pulmonary fibrosis, AEC mtDNA damage and apoptosis following exposure to asbestos or bleomycin suggests an important role for AEC mitochondrial H2O2-induced mtDNA damage in promoting lung fibrosis. We reason that strategies aimed at limiting AEC mtDNA damage arising from excess mitochondrial H2O2 production may be a novel therapeutic target for mitigating pulmonary fibrosis.
Collapse
Affiliation(s)
- Seok-Jo Kim
- Department of Medicine, Division of Pulmonary & Critical Care Medicine, Jesse Brown VA Medical Center, Chicago, IL, United States; Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States
| | - Paul Cheresh
- Department of Medicine, Division of Pulmonary & Critical Care Medicine, Jesse Brown VA Medical Center, Chicago, IL, United States; Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States
| | - Renea P Jablonski
- Department of Medicine, Division of Pulmonary & Critical Care Medicine, Jesse Brown VA Medical Center, Chicago, IL, United States; Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States
| | - Luisa Morales-Nebreda
- Department of Medicine, Division of Pulmonary & Critical Care Medicine, Jesse Brown VA Medical Center, Chicago, IL, United States; Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States
| | - Yuan Cheng
- Department of Medicine, Division of Pulmonary & Critical Care Medicine, Jesse Brown VA Medical Center, Chicago, IL, United States; Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States
| | - Erin Hogan
- Department of Medicine, Division of Pulmonary & Critical Care Medicine, Jesse Brown VA Medical Center, Chicago, IL, United States; Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States
| | - Anjana Yeldandi
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States
| | - Monica Chi
- Department of Medicine, Division of Pulmonary & Critical Care Medicine, Jesse Brown VA Medical Center, Chicago, IL, United States; Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States
| | - Raul Piseaux
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States
| | - Karen Ridge
- Department of Medicine, Division of Pulmonary & Critical Care Medicine, Jesse Brown VA Medical Center, Chicago, IL, United States; Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States
| | - C Michael Hart
- Atlanta VA Medical Center, Decatur, GA, United States; Department of Medicine, Emory University, Atlanta, GA, United States
| | - Navdeep Chandel
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States
| | - G R Scott Budinger
- Department of Medicine, Division of Pulmonary & Critical Care Medicine, Jesse Brown VA Medical Center, Chicago, IL, United States; Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States
| | - David W Kamp
- Department of Medicine, Division of Pulmonary & Critical Care Medicine, Jesse Brown VA Medical Center, Chicago, IL, United States; Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States.
| |
Collapse
|
65
|
Lee SH, Dudok B, Parihar VK, Jung KM, Zöldi M, Kang YJ, Maroso M, Alexander AL, Nelson GA, Piomelli D, Katona I, Limoli CL, Soltesz I. Neurophysiology of space travel: energetic solar particles cause cell type-specific plasticity of neurotransmission. Brain Struct Funct 2016; 222:2345-2357. [PMID: 27905022 PMCID: PMC5504243 DOI: 10.1007/s00429-016-1345-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 11/25/2016] [Indexed: 12/03/2022]
Abstract
In the not too distant future, humankind will embark on one of its greatest adventures, the travel to distant planets. However, deep space travel is associated with an inevitable exposure to radiation fields. Space-relevant doses of protons elicit persistent disruptions in cognition and neuronal structure. However, whether space-relevant irradiation alters neurotransmission is unknown. Within the hippocampus, a brain region crucial for cognition, perisomatic inhibitory control of pyramidal cells (PCs) is supplied by two distinct cell types, the cannabinoid type 1 receptor (CB1)-expressing basket cells (CB1BCs) and parvalbumin (PV)-expressing interneurons (PVINs). Mice subjected to low-dose proton irradiation were analyzed using electrophysiological, biochemical and imaging techniques months after exposure. In irradiated mice, GABA release from CB1BCs onto PCs was dramatically increased. This effect was abolished by CB1 blockade, indicating that irradiation decreased CB1-dependent tonic inhibition of GABA release. These alterations in GABA release were accompanied by decreased levels of the major CB1 ligand 2-arachidonoylglycerol. In contrast, GABA release from PVINs was unchanged, and the excitatory connectivity from PCs to the interneurons also underwent cell type-specific alterations. These results demonstrate that energetic charged particles at space-relevant low doses elicit surprisingly selective long-term plasticity of synaptic microcircuits in the hippocampus. The magnitude and persistent nature of these alterations in synaptic function are consistent with the observed perturbations in cognitive performance after irradiation, while the high specificity of these changes indicates that it may be possible to develop targeted therapeutic interventions to decrease the risk of adverse events during interplanetary travel.
Collapse
Affiliation(s)
- Sang-Hun Lee
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, 92697, USA. .,Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA.
| | - Barna Dudok
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, 1083, Budapest, Hungary.,School of Ph.D. Studies, Semmelweis University, Budapest, Hungary
| | - Vipan K Parihar
- Department of Radiation Oncology, University of California, Irvine, CA, 92697, USA
| | - Kwang-Mook Jung
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, 92697, USA
| | - Miklós Zöldi
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, 1083, Budapest, Hungary.,School of Ph.D. Studies, Semmelweis University, Budapest, Hungary
| | - Young-Jin Kang
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Mattia Maroso
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, 92697, USA.,Department of Neurosurgery, and Neurology and Neurological Sciences, Stanford University, Palo Alto, CA, 94305, USA
| | - Allyson L Alexander
- Department of Neurosurgery, and Neurology and Neurological Sciences, Stanford University, Palo Alto, CA, 94305, USA
| | - Gregory A Nelson
- Division of Radiation Research, Department of Basic Sciences, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Daniele Piomelli
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, 92697, USA
| | - István Katona
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, 1083, Budapest, Hungary
| | - Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine, CA, 92697, USA
| | - Ivan Soltesz
- Department of Neurosurgery, and Neurology and Neurological Sciences, Stanford University, Palo Alto, CA, 94305, USA
| |
Collapse
|
66
|
Raber J, Davis MJ, Pfankuch T, Rosenthal R, Doctrow SR, Moulder JE. Mitigating effect of EUK-207 on radiation-induced cognitive impairments. Behav Brain Res 2016; 320:457-463. [PMID: 27789343 DOI: 10.1016/j.bbr.2016.10.038] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 10/21/2016] [Accepted: 10/23/2016] [Indexed: 10/20/2022]
Abstract
The brain could be exposed to irradiation as part of a nuclear accident, radiological terrorism (dirty bomb scenario) or a medical radiological procedure. In the context of accidents or terrorism, there is considerable interest in compounds that can mitigate radiation-induced injury when treatment is initiated a day or more after the radiation exposure. As it will be challenging to determine the radiation exposure an individual has received within a relatively short time frame, it is also critical that the mitigating agent does not negatively affect individuals, including emergency workers, who might be treated, but who were not exposed. Alterations in hippocampus-dependent cognition often characterize radiation-induced cognitive injury. The catalytic ROS scavenger EUK-207 is a member of the class of metal-containing salen manganese (Mn) complexes that suppress oxidative stress, including in the mitochondria, and have been shown to mitigate radiation dermatitis, promote wound healing in irradiated skin, and mitigate vascular injuries in irradiated lungs. As the effects of EUK-207 against radiation injury in the brain are not known, we assessed the effects of EUK-207 on sham-irradiated animals and the ability of EUK-207 to mitigate radiation-induced cognitive injury. The day following irradiation or sham-irradiation, the mice started to receive EUK-207 and were cognitively tested 3 months following exposure. Mice irradiated at a dose of 15Gy showed cognitive impairments in the water maze probe trial. EUK-207 mitigated these impairments while not affecting cognitive performance of sham-irradiated mice in the water maze probe trial. Thus, EUK-207 has attractive properties and should be considered an ideal candidate to mitigate radiation-induced cognitive injury.
Collapse
Affiliation(s)
- J Raber
- Department of Behavioral Neuroscience, L470, Oregon Health and Science University, Portland, OR 97239, USA; Departments of Neurology and Radiation Medicine, Division of Neuroscience, ONPRC, L470, Oregon Health and Science University, Portland, Oregon 97239, USA.
| | - M J Davis
- Department of Behavioral Neuroscience, L470, Oregon Health and Science University, Portland, OR 97239, USA
| | - T Pfankuch
- Department of Behavioral Neuroscience, L470, Oregon Health and Science University, Portland, OR 97239, USA
| | - R Rosenthal
- Pulmonary Center, Boston University School of Medicine, MA 02215, USA
| | - S R Doctrow
- Pulmonary Center, Boston University School of Medicine, MA 02215, USA
| | - J E Moulder
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
67
|
Betlazar C, Middleton RJ, Banati RB, Liu GJ. The impact of high and low dose ionising radiation on the central nervous system. Redox Biol 2016; 9:144-156. [PMID: 27544883 PMCID: PMC4993858 DOI: 10.1016/j.redox.2016.08.002] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 08/06/2016] [Accepted: 08/09/2016] [Indexed: 12/12/2022] Open
Abstract
Responses of the central nervous system (CNS) to stressors and injuries, such as ionising radiation, are modulated by the concomitant responses of the brains innate immune effector cells, microglia. Exposure to high doses of ionising radiation in brain tissue leads to the expression and release of biochemical mediators of ‘neuroinflammation’, such as pro-inflammatory cytokines and reactive oxygen species (ROS), leading to tissue destruction. Contrastingly, low dose ionising radiation may reduce vulnerability to subsequent exposure of ionising radiation, largely through the stimulation of adaptive responses, such as antioxidant defences. These disparate responses may be reflective of non-linear differential microglial activation at low and high doses, manifesting as an anti-inflammatory or pro-inflammatory functional state. Biomarkers of pathology in the brain, such as the mitochondrial Translocator Protein 18 kDa (TSPO), have facilitated in vivo characterisation of microglial activation and ‘neuroinflammation’ in many pathological states of the CNS, though the exact function of TSPO in these responses remains elusive. Based on the known responsiveness of TSPO expression to a wide range of noxious stimuli, we discuss TSPO as a potential biomarker of radiation-induced effects. Ionising radiation can modulate responses of microglial cells in the CNS. High doses can induce ROS formation, oxidative stress and neuroinflammation. Low doses can mitigate tissue damage via antioxidant defences. TSPO as a potential biomarker and modulator of radiation induced effects in the CNS. Non-linear differential microglial activation to high and low doses is proposed.
Collapse
Affiliation(s)
- Calina Betlazar
- Bioanalytics group, Life Sciences, Australian Nuclear Science and Technology Organisation (ANSTO), New Illawarra Road, Lucas Heights, NSW 2234, Australia; Discipline of Medical Imaging & Radiation Sciences, Faculty of Health Sciences, The University of Sydney, 75 East Street, Lidcombe, NSW 2141, Australia
| | - Ryan J Middleton
- Bioanalytics group, Life Sciences, Australian Nuclear Science and Technology Organisation (ANSTO), New Illawarra Road, Lucas Heights, NSW 2234, Australia
| | - Richard B Banati
- Bioanalytics group, Life Sciences, Australian Nuclear Science and Technology Organisation (ANSTO), New Illawarra Road, Lucas Heights, NSW 2234, Australia; Discipline of Medical Imaging & Radiation Sciences, Faculty of Health Sciences, The University of Sydney, 75 East Street, Lidcombe, NSW 2141, Australia.
| | - Guo-Jun Liu
- Bioanalytics group, Life Sciences, Australian Nuclear Science and Technology Organisation (ANSTO), New Illawarra Road, Lucas Heights, NSW 2234, Australia; Discipline of Medical Imaging & Radiation Sciences, Faculty of Health Sciences, The University of Sydney, 75 East Street, Lidcombe, NSW 2141, Australia.
| |
Collapse
|
68
|
Patel M. Targeting Oxidative Stress in Central Nervous System Disorders. Trends Pharmacol Sci 2016; 37:768-778. [PMID: 27491897 DOI: 10.1016/j.tips.2016.06.007] [Citation(s) in RCA: 198] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 06/08/2016] [Accepted: 06/10/2016] [Indexed: 12/12/2022]
Abstract
There is widespread recognition that reactive oxygen species (ROS) play key roles in normal brain function and pathology in the context of neurological disease. Oxidative stress continues to be a key therapeutic target for neurological diseases. In developing antioxidant therapies for neurological disease, special attention should be given to the brain's unique vulnerability to oxidative insults and its architecture. Consideration of antioxidant therapy should be guided by a strong rationale for oxidative stress in a given neurological disease. This review provides an overview of processes that can guide the development of antioxidant therapies in neurological diseases, such as knowledge of basic redox mechanisms, unique features of brain pathophysiology, mechanisms and classes of antioxidants, and desirable properties of drug candidates.
Collapse
Affiliation(s)
- Manisha Patel
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
69
|
Basisty N, Dai D, Gagnidze A, Gitari L, Fredrickson J, Maina Y, Beyer RP, Emond MJ, Hsieh EJ, MacCoss MJ, Martin GM, Rabinovitch PS. Mitochondrial-targeted catalase is good for the old mouse proteome, but not for the young: 'reverse' antagonistic pleiotropy? Aging Cell 2016; 15:634-45. [PMID: 27061426 PMCID: PMC4933659 DOI: 10.1111/acel.12472] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/22/2016] [Indexed: 12/31/2022] Open
Abstract
Reactive oxygen species (ROS) are highly reactive oxygen‐containing molecules associated with aging and a broad spectrum of pathologies. We have previously shown that transgenic expression of the antioxidant enzyme catalase targeted to the mitochondria (mCAT) in mice reduces ROS, attenuates age‐related disease, and increases lifespan. However, it has been increasingly recognized that ROS also has beneficial roles in signaling, hormesis, stress response, and immunity. We therefore hypothesized that mCAT might be beneficial only when ROS approaches pathological levels in older age and might not be advantageous at a younger age when basal ROS is low. We analyzed abundance and turnover of the global proteome in hearts and livers of young (4 month) and old (20 month) mCAT and wild‐type (WT) mice. In old hearts and livers of WT mice, protein half‐lives were reduced compared to young, while in mCAT mice the reverse was observed; the longest half‐lives were seen in old mCAT mice and the shortest in young mCAT. Protein abundance of old mCAT hearts recapitulated a more youthful proteomic expression profile (P‐value < 0.01). However, young mCAT mice partially phenocopied the older wild‐type proteome (P‐value < 0.01). Age strongly interacts with mCAT, consistent with antagonistic pleiotropy in the reverse of the typical direction. These findings underscore the contrasting roles of ROS in young vs. old mice and indicate the need for better understanding of the interaction between dose and age in assessing the efficacy of therapeutic interventions in aging, including mitochondrial antioxidants.
Collapse
Affiliation(s)
- Nathan Basisty
- Department of Pathology University of Washington 1959 NE Pacific Ave Seattle WA 98195 USA
| | - Dao‐Fu Dai
- Department of Pathology University of Washington 1959 NE Pacific Ave Seattle WA 98195 USA
| | - Arni Gagnidze
- Department of Pathology University of Washington 1959 NE Pacific Ave Seattle WA 98195 USA
| | - Lemuel Gitari
- Department of Pathology University of Washington 1959 NE Pacific Ave Seattle WA 98195 USA
| | - Jeanne Fredrickson
- Department of Pathology University of Washington 1959 NE Pacific Ave Seattle WA 98195 USA
| | - Yvonne Maina
- Department of Pathology University of Washington 1959 NE Pacific Ave Seattle WA 98195 USA
| | - Richard P. Beyer
- Department of Environmental Health University of Washington 1959 NE Pacific Ave Seattle WA 98195 USA
| | - Mary J. Emond
- Department of Biostatistics University of Washington 1959 NE Pacific Ave Seattle WA 98195 USA
| | - Edward J. Hsieh
- Department of Genome Sciences University of Washington 1959 NE Pacific Ave Seattle WA 98195 USA
| | - Michael J. MacCoss
- Department of Genome Sciences University of Washington 1959 NE Pacific Ave Seattle WA 98195 USA
| | - George M. Martin
- Department of Pathology University of Washington 1959 NE Pacific Ave Seattle WA 98195 USA
| | - Peter S. Rabinovitch
- Department of Pathology University of Washington 1959 NE Pacific Ave Seattle WA 98195 USA
| |
Collapse
|
70
|
Acharya MM, Baulch JE, Lusardi TA, Allen BD, Chmielewski NN, Baddour AAD, Limoli CL, Boison D. Adenosine Kinase Inhibition Protects against Cranial Radiation-Induced Cognitive Dysfunction. Front Mol Neurosci 2016; 9:42. [PMID: 27375429 PMCID: PMC4891332 DOI: 10.3389/fnmol.2016.00042] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 05/20/2016] [Indexed: 12/13/2022] Open
Abstract
Clinical radiation therapy for the treatment of CNS cancers leads to unintended and debilitating impairments in cognition. Radiation-induced cognitive dysfunction is long lasting; however, the underlying molecular and cellular mechanisms are still not well established. Since ionizing radiation causes microglial and astroglial activation, we hypothesized that maladaptive changes in astrocyte function might be implicated in radiation-induced cognitive dysfunction. Among other gliotransmitters, astrocytes control the availability of adenosine, an endogenous neuroprotectant and modulator of cognition, via metabolic clearance through adenosine kinase (ADK). Adult rats exposed to cranial irradiation (10 Gy) showed significant declines in performance of hippocampal-dependent cognitive function tasks [novel place recognition, novel object recognition (NOR), and contextual fear conditioning (FC)] 1 month after exposure to ionizing radiation using a clinically relevant regimen. Irradiated rats spent less time exploring a novel place or object. Cranial irradiation also led to reduction in freezing behavior compared to controls in the FC task. Importantly, immunohistochemical analyses of irradiated brains showed significant elevation of ADK immunoreactivity in the hippocampus that was related to astrogliosis and increased expression of glial fibrillary acidic protein (GFAP). Conversely, rats treated with the ADK inhibitor 5-iodotubercidin (5-ITU, 3.1 mg/kg, i.p., for 6 days) prior to cranial irradiation showed significantly improved behavioral performance in all cognitive tasks 1 month post exposure. Treatment with 5-ITU attenuated radiation-induced astrogliosis and elevated ADK immunoreactivity in the hippocampus. These results confirm an astrocyte-mediated mechanism where preservation of extracellular adenosine can exert neuroprotection against radiation-induced pathology. These innovative findings link radiation-induced changes in cognition and CNS functionality to altered purine metabolism and astrogliosis, thereby linking the importance of adenosine homeostasis in the brain to radiation injury.
Collapse
Affiliation(s)
- Munjal M Acharya
- Department of Radiation Oncology, University of California Irvine, CA, USA
| | - Janet E Baulch
- Department of Radiation Oncology, University of California Irvine, CA, USA
| | - Theresa A Lusardi
- R. S. Dow Neurobiology Laboratories, Legacy Research Institute Portland, OR, USA
| | - Barrett D Allen
- Department of Radiation Oncology, University of California Irvine, CA, USA
| | | | - Al Anoud D Baddour
- Department of Radiation Oncology, University of California Irvine, CA, USA
| | - Charles L Limoli
- Department of Radiation Oncology, University of California Irvine, CA, USA
| | - Detlev Boison
- R. S. Dow Neurobiology Laboratories, Legacy Research Institute Portland, OR, USA
| |
Collapse
|
71
|
Chmielewski NN, Caressi C, Giedzinski E, Parihar VK, Limoli CL. Contrasting the effects of proton irradiation on dendritic complexity of subiculum neurons in wild type and MCAT mice. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2016; 57:364-371. [PMID: 26996825 DOI: 10.1002/em.22006] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 02/19/2016] [Indexed: 06/05/2023]
Abstract
Growing evidence suggests that radiation-induced oxidative stress directly affects a wide range of biological changes with an overall negative impact on CNS function. In the past we have demonstrated that transgenic mice over-expressing human catalase targeted to the mitochondria (MCAT) exhibit a range of neuroprotective phenotypes following irradiation that include improved neurogenesis, dendritic complexity, and cognition. To determine the extent of the neuroprotective phenotype afforded by MCAT expression in different hippocampal regions, we analyzed subiculum neurons for changes in neuronal structure and synaptic integrity after exposure to low dose (0.5 Gy) 150 MeV proton irradiation. One month following irradiation of WT and MCAT mice, a range of morphometric parameters were quantified along Golgi-Cox impregnated neurons. Compared with WT mice, subiculum neurons from MCAT mice exhibited increased trends (albeit not statistically significant) toward increased dendritic complexity in both control and irradiated cohorts. However, Sholl analysis of MCAT mice revealed significantly increased arborization of the distal dendritic tree, indicating a protective effect on secondary and tertiary branching. Interestingly, radiation-induced increases in postsynaptic density protein (PSD-95) puncta were not as pronounced in MCAT compared with WT mice, and were significantly lower after the 0.5 Gy dose. As past data has linked radiation exposure to reduced dendritic complexity, elevated PSD-95 and impaired cognition, reductions in mitochondrial oxidative stress have proven useful in ameliorating many of these radiation-induced sequelae. Data presented here shows similar trends, and again points to the potential benefits of reducing oxidative stress in the brain to attenuate radiation injury. Environ. Mol. Mutagen. 57:364-371, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | - Chongshan Caressi
- Department of Radiation Oncology, University of California, Irvine, California
| | - Erich Giedzinski
- Department of Radiation Oncology, University of California, Irvine, California
| | - Vipan K Parihar
- Department of Radiation Oncology, University of California, Irvine, California
| | - Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine, California
| |
Collapse
|
72
|
Limoli CL. Understanding and targeting dynamic stress responses of the brain: What we have learned and how to improve neurocognitive outcome following neurotoxic insult. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2016; 57:319-321. [PMID: 27208487 DOI: 10.1002/em.22022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 04/28/2016] [Indexed: 06/05/2023]
Affiliation(s)
- Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine, California
| |
Collapse
|
73
|
Sridharan DM, Asaithamby A, Blattnig SR, Costes SV, Doetsch PW, Dynan WS, Hahnfeldt P, Hlatky L, Kidane Y, Kronenberg A, Naidu MD, Peterson LE, Plante I, Ponomarev AL, Saha J, Snijders AM, Srinivasan K, Tang J, Werner E, Pluth JM. Evaluating biomarkers to model cancer risk post cosmic ray exposure. LIFE SCIENCES IN SPACE RESEARCH 2016; 9:19-47. [PMID: 27345199 PMCID: PMC5613937 DOI: 10.1016/j.lssr.2016.05.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 05/11/2016] [Indexed: 06/06/2023]
Abstract
Robust predictive models are essential to manage the risk of radiation-induced carcinogenesis. Chronic exposure to cosmic rays in the context of the complex deep space environment may place astronauts at high cancer risk. To estimate this risk, it is critical to understand how radiation-induced cellular stress impacts cell fate decisions and how this in turn alters the risk of carcinogenesis. Exposure to the heavy ion component of cosmic rays triggers a multitude of cellular changes, depending on the rate of exposure, the type of damage incurred and individual susceptibility. Heterogeneity in dose, dose rate, radiation quality, energy and particle flux contribute to the complexity of risk assessment. To unravel the impact of each of these factors, it is critical to identify sensitive biomarkers that can serve as inputs for robust modeling of individual risk of cancer or other long-term health consequences of exposure. Limitations in sensitivity of biomarkers to dose and dose rate, and the complexity of longitudinal monitoring, are some of the factors that increase uncertainties in the output from risk prediction models. Here, we critically evaluate candidate early and late biomarkers of radiation exposure and discuss their usefulness in predicting cell fate decisions. Some of the biomarkers we have reviewed include complex clustered DNA damage, persistent DNA repair foci, reactive oxygen species, chromosome aberrations and inflammation. Other biomarkers discussed, often assayed for at longer points post exposure, include mutations, chromosome aberrations, reactive oxygen species and telomere length changes. We discuss the relationship of biomarkers to different potential cell fates, including proliferation, apoptosis, senescence, and loss of stemness, which can propagate genomic instability and alter tissue composition and the underlying mRNA signatures that contribute to cell fate decisions. Our goal is to highlight factors that are important in choosing biomarkers and to evaluate the potential for biomarkers to inform models of post exposure cancer risk. Because cellular stress response pathways to space radiation and environmental carcinogens share common nodes, biomarker-driven risk models may be broadly applicable for estimating risks for other carcinogens.
Collapse
Affiliation(s)
| | | | - Steve R Blattnig
- Langley Research Center, Langley Research Center (LaRC), VA, United States
| | - Sylvain V Costes
- Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| | | | | | | | - Lynn Hlatky
- CCSB-Tufts School of Medicine, Boston, MA, United States
| | - Yared Kidane
- Wyle Science, Technology & Engineering Group, Houston, TX, United States
| | - Amy Kronenberg
- Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| | - Mamta D Naidu
- CCSB-Tufts School of Medicine, Boston, MA, United States
| | - Leif E Peterson
- Houston Methodist Research Institute, Houston, TX, United States
| | - Ianik Plante
- Wyle Science, Technology & Engineering Group, Houston, TX, United States
| | - Artem L Ponomarev
- Wyle Science, Technology & Engineering Group, Houston, TX, United States
| | - Janapriya Saha
- UT Southwestern Medical Center, Dallas, TX, United States
| | | | | | - Jonathan Tang
- Exogen Biotechnology, Inc., Berkeley, CA, United States
| | | | - Janice M Pluth
- Lawrence Berkeley National Laboratory, Berkeley, CA, United States.
| |
Collapse
|
74
|
Allen AR, Raber J, Chakraborti A, Sharma S, Fike JR. 56Fe Irradiation Alters Spine Density and Dendritic Complexity in the Mouse Hippocampus. Radiat Res 2015; 184:586-94. [DOI: 10.1667/rr14103.1] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
75
|
Tomé WA, Gökhan Ş, Gulinello ME, Brodin NP, Heard J, Mehler MF, Guha C. Hippocampal-dependent neurocognitive impairment following cranial irradiation observed in pre-clinical models: current knowledge and possible future directions. Br J Radiol 2015; 89:20150762. [PMID: 26514377 DOI: 10.1259/bjr.20150762] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
We reviewed the literature for studies pertaining to impaired adult neurogenesis leading to neurocognitive impairment following cranial irradiation in rodent models. This compendium was compared with respect to radiation dose, converted to equivalent dose in 2 Gy fractions (EQD2) to allow for direct comparison between studies. The effects of differences between animal species and the dependence on animal age as well as for time after irradiation were also considered. One of the major sites of de novo adult neurogenesis is the hippocampus, and as such, this review also focuses on assessing evidence related to the expression and potential effects of inflammatory cytokines on neural stem cells in the subgranular zone of the dentate gyrus and whether this correlates with neurocognitive impairment. This review also discusses potential strategies to mitigate the detrimental effects on neurogenesis and neurocognition resulting from cranial irradiation, and how the rationale for these strategies compares with the current outcome of pre-clinical studies.
Collapse
Affiliation(s)
- Wolfgang A Tomé
- 1 Institute for Onco-Physics, Department of Radiation Oncology, Albert Einstein College of Medicine, Bronx, NY, USA.,2 Department of Radiation Oncology, Montefiore Medical Center, Bronx, NY, USA.,3 Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Şölen Gökhan
- 3 Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Maria E Gulinello
- 4 Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - N Patrik Brodin
- 1 Institute for Onco-Physics, Department of Radiation Oncology, Albert Einstein College of Medicine, Bronx, NY, USA.,2 Department of Radiation Oncology, Montefiore Medical Center, Bronx, NY, USA
| | - John Heard
- 2 Department of Radiation Oncology, Montefiore Medical Center, Bronx, NY, USA
| | - Mark F Mehler
- 3 Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, USA.,4 Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA.,5 Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Chandan Guha
- 1 Institute for Onco-Physics, Department of Radiation Oncology, Albert Einstein College of Medicine, Bronx, NY, USA.,2 Department of Radiation Oncology, Montefiore Medical Center, Bronx, NY, USA
| |
Collapse
|
76
|
Bellone JA, Rudobeck E, Hartman RE, Szücs A, Vlkolinský R. A Single Low Dose of Proton Radiation Induces Long-Term Behavioral and Electrophysiological Changes in Mice. Radiat Res 2015. [DOI: 10.1667/rr13903.1] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
77
|
Parihar VK, Allen B, Tran KK, Macaraeg TG, Chu EM, Kwok SF, Chmielewski NN, Craver BM, Baulch JE, Acharya MM, Cucinotta FA, Limoli CL. What happens to your brain on the way to Mars. SCIENCE ADVANCES 2015; 1:e1400256. [PMID: 26180843 PMCID: PMC4500198 DOI: 10.1126/sciadv.1400256] [Citation(s) in RCA: 145] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 04/05/2015] [Indexed: 05/21/2023]
Abstract
As NASA prepares for the first manned spaceflight to Mars, questions have surfaced concerning the potential for increased risks associated with exposure to the spectrum of highly energetic nuclei that comprise galactic cosmic rays. Animal models have revealed an unexpected sensitivity of mature neurons in the brain to charged particles found in space. Astronaut autonomy during long-term space travel is particularly critical as is the need to properly manage planned and unanticipated events, activities that could be compromised by accumulating particle traversals through the brain. Using mice subjected to space-relevant fluences of charged particles, we show significant cortical- and hippocampal-based performance decrements 6 weeks after acute exposure. Animals manifesting cognitive decrements exhibited marked and persistent radiation-induced reductions in dendritic complexity and spine density along medial prefrontal cortical neurons known to mediate neurotransmission specifically interrogated by our behavioral tasks. Significant increases in postsynaptic density protein 95 (PSD-95) revealed major radiation-induced alterations in synaptic integrity. Impaired behavioral performance of individual animals correlated significantly with reduced spine density and trended with increased synaptic puncta, thereby providing quantitative measures of risk for developing cognitive decrements. Our data indicate an unexpected and unique susceptibility of the central nervous system to space radiation exposure, and argue that the underlying radiation sensitivity of delicate neuronal structure may well predispose astronauts to unintended mission-critical performance decrements and/or longer-term neurocognitive sequelae.
Collapse
Affiliation(s)
- Vipan K. Parihar
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA 92697–2695, USA
| | - Barrett Allen
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA 92697–2695, USA
| | - Katherine K. Tran
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA 92697–2695, USA
| | - Trisha G. Macaraeg
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA 92697–2695, USA
| | - Esther M. Chu
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA 92697–2695, USA
| | - Stephanie F. Kwok
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA 92697–2695, USA
| | - Nicole N. Chmielewski
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA 92697–2695, USA
| | - Brianna M. Craver
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA 92697–2695, USA
| | - Janet E. Baulch
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA 92697–2695, USA
| | - Munjal M. Acharya
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA 92697–2695, USA
| | - Francis A. Cucinotta
- Department of Health Physics and Diagnostic Sciences, University of Nevada, Las Vegas, Las Vegas, NV 89154, USA
| | - Charles L. Limoli
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA 92697–2695, USA
- Corresponding author. E-mail:
| |
Collapse
|
78
|
Baulch JE, Craver BM, Tran KK, Yu L, Chmielewski N, Allen BD, Limoli CL. Persistent oxidative stress in human neural stem cells exposed to low fluences of charged particles. Redox Biol 2015; 5:24-32. [PMID: 25800120 PMCID: PMC4371546 DOI: 10.1016/j.redox.2015.03.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 03/10/2015] [Indexed: 10/30/2022] Open
Abstract
Exposure to the space radiation environment poses risks for a range of deleterious health effects due to the unique types of radiation encountered. Galactic cosmic rays are comprised of a spectrum of highly energetic nuclei that deposit densely ionizing tracks of damage along the particle trajectory. These tracks are distinct from those generated by the more sparsely ionizing terrestrial radiations, and define the geometric distribution of the complex cellular damage that results when charged particles traverse the tissues of the body. The exquisite radiosensitivity of multipotent neural stem and progenitor cells found within the neurogenic regions of the brain predispose the central nervous system to elevated risks for radiation induced sequelae. Here we show that human neural stem cells (hNSC) exposed to different charged particles at space relevant fluences exhibit significant and persistent oxidative stress. Radiation induced oxidative stress was found to be most dependent on total dose rather than on the linear energy transfer of the incident particle. The use of redox sensitive fluorogenic dyes possessing relative specificity for hydroxyl radicals, peroxynitrite, nitric oxide (NO) and mitochondrial superoxide confirmed that most irradiation paradigms elevated reactive oxygen and nitrogen species (ROS and RNS, respectively) in hNSC over a 1 week interval following exposure. Nitric oxide synthase (NOS) was not the major source of elevated nitric oxides, as the use of NOS inhibitors had little effect on NO dependent fluorescence. Our data provide extensive evidence for the capability of low doses of charged particles to elicit marked changes in the metabolic profile of irradiated hNSC. Radiation induced changes in redox state may render the brain more susceptible to the development of neurocognitive deficits that could affect an astronaut's ability to perform complex tasks during extended missions in deep space.
Collapse
Affiliation(s)
- Janet E Baulch
- Department of Radiation Oncology, University of California, Irvine, CA 92697-2695, USA
| | - Brianna M Craver
- Department of Radiation Oncology, University of California, Irvine, CA 92697-2695, USA
| | - Katherine K Tran
- Department of Radiation Oncology, University of California, Irvine, CA 92697-2695, USA
| | - Liping Yu
- Department of Radiation Oncology, University of California, Irvine, CA 92697-2695, USA
| | - Nicole Chmielewski
- Department of Radiation Oncology, University of California, Irvine, CA 92697-2695, USA
| | - Barrett D Allen
- Department of Radiation Oncology, University of California, Irvine, CA 92697-2695, USA
| | - Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine, CA 92697-2695, USA.
| |
Collapse
|