1
|
Cini N, Atasoy Ö, Uyanikgil Y, Yaprak G, Erdoğan MA, Erbas O. Ceftriaxone has a neuroprotective effect in a whole-brain irradiation-induced neurotoxicity model by increasing GLT-1 and reducing oxidative stress. Strahlenther Onkol 2025:10.1007/s00066-025-02405-z. [PMID: 40353856 DOI: 10.1007/s00066-025-02405-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 03/30/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND Radiation-induced brain injury is a prominent side effect of whole-brain irradiation (IR) due to triggered oxidative and inflammatory processes, often resulting in severe and debilitating cognitive dysfunction and neuronal damage. The development of persistent oxidative stress results from radiation-induced reactive oxygen species. Another result is the initiation of glutamate excitotoxicity, which is closely associated with changes in glutamate levels. Elevated release or reduced glutamate uptake disrupts neuronal homeostasis, leading to oxidative stress, mitochondrial dysfunction, and neuroinflammation. The neuroprotective and antioxidant properties of ceftriaxone (CTX) have been linked to its ability to reduce glutamate excitotoxicity, inflammation, and to modulate oxidative stress. MATERIALS AND METHODS Twenty-one female Wistar rats were included in the study, and 14 of them underwent whole-brain IR with a single dose of 20 Gy on day 7. Saline and CTX applications continued for 21 days. The animals were divided into three groups: group 1: normal control; group 2: IR + saline; and group 3: IR + CTX. To compare the groups, a one-way analysis of variance (ANOVA) statistical test was employed, with a significance threshold set at p < 0.05. RESULTS Ceftriaxone treatment had a positive impact on the results of various assessments, e.g., behavioral tests including the three-chamber sociability test, the open-field test, and passive avoidance learning. It also led to increased counts of hippocampal CA1, CA3, and Purkinje neurons as well as elevated brain levels of brain-derived neurotrophic factor (BDNF), glutamate transporter 1 (GLT-1), and superoxide dismutase (SOD) activity. Conversely, CTX reduced the glial fibrillary acidic protein (GFAP) immunostaining index as well as brain levels of malondialdehyde (MDA) and tumor necrosis factor alpha (TNF-α). CONCLUSION Ceftriaxone demonstrated promising effectiveness in mitigating radiation-induced neurocognitive impairments and the deterioration of social memory capacity. This effect is achieved by reducing neuronal loss, oxidative stress, and neuroinflammation in irradiated rat brains. Furthermore, the application of CTX facilitated removal of excess glutamate from synapses, thus preventing glutamate excitotoxicity and protecting neurons from excitotoxic cell death.
Collapse
Affiliation(s)
- Nilsu Cini
- Department of Radiation Oncology, Kartal Dr. Lütfi Kırdar City Hospital, Istanbul, Turkey.
| | - Özüm Atasoy
- Department of Radiation Oncology, Giresun Education and Research Hospital, Giresun, Turkey
- Department of Medical Biochemistry, Istanbul Medeniyet University, Faculty of Medicine, Istanbul, Turkey
| | - Yigit Uyanikgil
- Department of Histology and Embryology, Ege University, Faculty of Medicine, Izmir, Turkey
| | - Gökhan Yaprak
- Department of Radiation Oncology, Kartal Dr. Lütfi Kırdar City Hospital, Istanbul, Turkey
| | - Mümin Alper Erdoğan
- Department of Physiology, Izmir Katip Celebi University, Faculty of Medicine, Izmir, Turkey
| | - Oytun Erbas
- Department of Physiology, Demiroğlu Bilim University, Faculty of Medicine, Istanbul, Turkey
| |
Collapse
|
2
|
Leskinen S, Alsalek S, Wernicke AG. Effects of radiotherapy on the hippocampus and hippocampal neurogenesis: a systematic review of preclinical studies. Strahlenther Onkol 2025; 201:383-397. [PMID: 39800777 DOI: 10.1007/s00066-024-02341-4] [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: 07/27/2024] [Accepted: 11/18/2024] [Indexed: 03/22/2025]
Abstract
PURPOSE A comprehensive literature review was undertaken to understand the effects and underlying mechanisms of cranial radiotherapy (RT) on the hippocampus and hippocampal neurogenesis as well as to explore protective factors and treatments that might mitigate these effects in preclinical studies. METHODS PubMed/MEDLINE, Web of Science, and Embase were queried for studies involving the effects of radiation on the hippocampus and hippocampal neurogenesis. Data extraction followed the Animal Research Reporting of In Vivo Experiments (ARRIVE) guidelines, and a risk of bias assessment was conducted for the included animal studies using the Systematic Review Centre for Laboratory Animal Experimentation (SYRCLE) risk of bias tool. RESULTS Ninety studies were included, with 48 assessing radiation-induced changes and 42 examining possible interventions. The majority of studies (97.8%) used experimental animal models. Studies demonstrated that cranial irradiation reduces hippocampal neurogenesis, particularly in the neurogenic niches of the dentate gyrus; causes alterations in gene expression and enzymatic activity; induces inflammation; promotes apoptosis; and often results in cognitive impairment. Potential protective strategies include pharmacological agents like metformin and peroxisome proliferator-activated receptor-α (PPAR-α) agonists or behavioral interventions like voluntary running. In a risk of bias assessment, many studies were rated as having an unclear risk of bias. CONCLUSION Radiotherapy, while essential for managing brain tumors, can have adverse effects on hippocampal function and structure in animal models. These effects manifest in reduced neurogenesis, molecular alterations, and increased inflammation, leading to cognitive deficits. Further research is needed to identify and improve interventions and develop comprehensive therapeutic approaches that balance effective tumor control with the preservation of cognitive health.
Collapse
Affiliation(s)
- Sandra Leskinen
- State University of New York Downstate Medical Center, Brooklyn, NY, USA.
| | - Samir Alsalek
- Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, CA, USA
| | - A Gabriella Wernicke
- Department of Radiation Medicine, Lenox Hill Hospital, Zucker School of Medicine at Hofstra/Northwell, New York, NY, USA
| |
Collapse
|
3
|
Lan J, Ren Y, Liu Y, Chen L, Liu J. A bibliometric analysis of radiation-induced brain injury: a research of the literature from 1998 to 2023. Discov Oncol 2024; 15:364. [PMID: 39172266 PMCID: PMC11341524 DOI: 10.1007/s12672-024-01223-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 08/06/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND Radiation-induced brain injury (RIBI) is a debilitating sequela after cranial radiotherapy. Research on the topic of RIBI has gradually entered the public eye, with more innovations and applications of evidence-based research and biological mechanism research in the field of that. This was the first bibliometric analysis on RIBI, assessing brain injury related to radiation articles that were published during 1998-2023, to provide an emerging theoretical basis for the future development of RIBI. METHODS Literature were obtained from the Web of Science Core Collection (WOSCC) from its inception to December 31, 2023. The column of publications, author details, affiliated institutions and countries, publication year, and keywords were also recorded. RESULTS A total of 2543 journal articles were selected. The annual publications on RIBI fluctuated within a certain range. Journal of Neuro-oncology was the most published journal and Radiation Oncology was the most impactful one. LIMOLI CL was the most prolific author with 37 articles and shared the highest h-index with BARNETT GH. The top one country and institutions were the USA and the University of California System, respectively. Clusters analysis of co-keywords demonstrated that the temporal research trends in this field primarily focused on imaging examination and therapy for RIBI. CONCLUSION This study collects, visualizes, and analyzes the literature within the field of RIBI over the last 25 years to map the development process, research frontiers and hotspots, and cutting-edge directions in clinical practice and mechanisms related to RIBI.
Collapse
Affiliation(s)
- Jinxin Lan
- Department of Neurosurgery, The First Medical Center, The Chinese PLA General Hospital, Beijing, 100853, China
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Yifan Ren
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Yuyang Liu
- Department of Neurosurgery, The 920th Hospital of Joint Logistics Support Force, Kunming, 650032, Yunnan, China
| | - Ling Chen
- Department of Neurosurgery, The First Medical Center, The Chinese PLA General Hospital, Beijing, 100853, China.
- Chinese PLA General Hospital, Chinese PLA Institute of Neurosurgery, 28 Fuxing Road, Haidian District, Beijing, 100853, China.
| | - Jialin Liu
- Department of Neurosurgery, The First Medical Center, The Chinese PLA General Hospital, Beijing, 100853, China.
- Chinese PLA General Hospital, Chinese PLA Institute of Neurosurgery, 28 Fuxing Road, Haidian District, Beijing, 100853, China.
| |
Collapse
|
4
|
Impey S, Raber J. Irradiation and Alterations in Hippocampal DNA Methylation. EPIGENOMES 2024; 8:27. [PMID: 39051185 PMCID: PMC11270359 DOI: 10.3390/epigenomes8030027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 06/11/2024] [Accepted: 07/02/2024] [Indexed: 07/27/2024] Open
Abstract
The response of the brain to radiation is important for cancer patients receiving whole or partial brain irradiation or total body irradiation, those exposed to irradiation as part of a nuclear accident or a nuclear war or terrorism event, and for astronauts during and following space missions. The mechanisms mediating the effects of irradiation on the hippocampus might be associated with alterations in hippocampal DNA methylation. Changes in cytosine methylation involving the addition of a methyl group to cytosine (5 mC) and especially those involving the addition of a hydroxy group to 5 mC (hydroxymethylcytosine or 5 hmC) play a key role in regulating the expression of genes required for hippocampal function. In this review article, we will discuss the effects of radiation on hippocampal DNA methylation and whether these effects are associated with hippocampus-dependent cognitive measures and molecular measures in the hippocampus involved in cognitive measures. We will also discuss whether the radiation-induced changes in hippocampal DNA methylation show an overlap across different doses of heavy ion irradiation and across irradiation with different ions. We will also discuss whether the DNA methylation changes show a tissue-dependent response.
Collapse
Affiliation(s)
- Soren Impey
- Dow Neurobiology Laboratories, Legacy Research Institute Legacy Health Systems, 1225 NE 2nd Ave, Portland, OR 97232, USA
- Departments of Behavioral Neuroscience, Neurology, and Radiation Medicine, Division of Neuroscience, ONPRC, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Jacob Raber
- Departments of Behavioral Neuroscience, Neurology, and Radiation Medicine, Division of Neuroscience, ONPRC, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| |
Collapse
|
5
|
Elajaili HB, Dee NM, Dikalov SI, Kao JPY, Nozik ES. Use of Electron Paramagnetic Resonance (EPR) to Evaluate Redox Status in a Preclinical Model of Acute Lung Injury. Mol Imaging Biol 2024; 26:495-502. [PMID: 37193807 PMCID: PMC10188229 DOI: 10.1007/s11307-023-01826-5] [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: 12/28/2022] [Revised: 05/03/2023] [Accepted: 05/08/2023] [Indexed: 05/18/2023]
Abstract
PURPOSE Patients with hyper- vs. hypo-inflammatory subphenotypes of acute respiratory distress syndrome (ARDS) exhibit different clinical outcomes. Inflammation increases the production of reactive oxygen species (ROS) and increased ROS contributes to the severity of illness. Our long-term goal is to develop electron paramagnetic resonance (EPR) imaging of lungs in vivo to precisely measure superoxide production in ARDS in real time. As a first step, this requires the development of in vivo EPR methods for quantifying superoxide generation in the lung during injury, and testing if such superoxide measurements can differentiate between susceptible and protected mouse strains. PROCEDURES In WT mice, mice lacking total body extracellular superoxide dismutase (EC-SOD) (KO), or mice overexpressing lung EC-SOD (Tg), lung injury was induced with intraperitoneal (IP) lipopolysaccharide (LPS) (10 mg/kg). At 24 h after LPS treatment, mice were injected with the cyclic hydroxylamines 1-hydroxy-3-carboxy-2,2,5,5-tetramethylpyrrolidine hydrochloride (CPH) or 4-acetoxymethoxycarbonyl-1-hydroxy-2,2,5,5-tetramethylpyrrolidine-3-carboxylic acid (DCP-AM-H) probes to detect, respectively, cellular and mitochondrial ROS - specifically superoxide. Several probe delivery strategies were tested. Lung tissue was collected up to one hour after probe administration and assayed by EPR. RESULTS As measured by X-band EPR, cellular and mitochondrial superoxide increased in the lungs of LPS-treated mice compared to control. Lung cellular superoxide was increased in EC-SOD KO mice and decreased in EC-SOD Tg mice compared to WT. We also validated an intratracheal (IT) delivery method, which enhanced the lung signal for both spin probes compared to IP administration. CONCLUSIONS We have developed protocols for delivering EPR spin probes in vivo, allowing detection of cellular and mitochondrial superoxide in lung injury by EPR. Superoxide measurements by EPR could differentiate mice with and without lung injury, as well as mouse strains with different disease susceptibilities. We expect these protocols to capture real-time superoxide production and enable evaluation of lung EPR imaging as a potential clinical tool for subphenotyping ARDS patients based on redox status.
Collapse
Affiliation(s)
- Hanan B Elajaili
- Pediatric Critical Care Medicine, University of Colorado Anschutz Medical Campus, 12700 E. 19th Ave., B131, Aurora, CO, 80045, USA
| | - Nathan M Dee
- Pediatric Critical Care Medicine, University of Colorado Anschutz Medical Campus, 12700 E. 19th Ave., B131, Aurora, CO, 80045, USA
| | - Sergey I Dikalov
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Joseph P Y Kao
- Center for Biomedical Engineering and Technology, and Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Eva S Nozik
- Pediatric Critical Care Medicine, University of Colorado Anschutz Medical Campus, 12700 E. 19th Ave., B131, Aurora, CO, 80045, USA.
| |
Collapse
|
6
|
Miller KB, Mi KL, Nelson GA, Norman RB, Patel ZS, Huff JL. Ionizing radiation, cerebrovascular disease, and consequent dementia: A review and proposed framework relevant to space radiation exposure. Front Physiol 2022; 13:1008640. [PMID: 36388106 PMCID: PMC9640983 DOI: 10.3389/fphys.2022.1008640] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 09/29/2022] [Indexed: 09/05/2023] Open
Abstract
Space exploration requires the characterization and management or mitigation of a variety of human health risks. Exposure to space radiation is one of the main health concerns because it has the potential to increase the risk of cancer, cardiovascular disease, and both acute and late neurodegeneration. Space radiation-induced decrements to the vascular system may impact the risk for cerebrovascular disease and consequent dementia. These risks may be independent or synergistic with direct damage to central nervous system tissues. The purpose of this work is to review epidemiological and experimental data regarding the impact of low-to-moderate dose ionizing radiation on the central nervous system and the cerebrovascular system. A proposed framework outlines how space radiation-induced effects on the vasculature may increase risk for both cerebrovascular dysfunction and neural and cognitive adverse outcomes. The results of this work suggest that there are multiple processes by which ionizing radiation exposure may impact cerebrovascular function including increases in oxidative stress, neuroinflammation, endothelial cell dysfunction, arterial stiffening, atherosclerosis, and cerebral amyloid angiopathy. Cerebrovascular adverse outcomes may also promote neural and cognitive adverse outcomes. However, there are many gaps in both the human and preclinical evidence base regarding the long-term impact of ionizing radiation exposure on brain health due to heterogeneity in both exposures and outcomes. The unique composition of the space radiation environment makes the translation of the evidence base from terrestrial exposures to space exposures difficult. Additional investigation and understanding of the impact of low-to-moderate doses of ionizing radiation including high (H) atomic number (Z) and energy (E) (HZE) ions on the cerebrovascular system is needed. Furthermore, investigation of how decrements in vascular systems may contribute to development of neurodegenerative diseases in independent or synergistic pathways is important for protecting the long-term health of astronauts.
Collapse
Affiliation(s)
| | | | - Gregory A. Nelson
- Department of Basic Sciences, Division of Biomedical Engineering Sciences, Loma Linda University, Loma Linda, CA, United States
- NASA Johnson Space Center, Houston, TX, United States
- KBR Inc., Houston, TX, United States
| | - Ryan B. Norman
- NASA Langley Research Center, Hampton, VA, United States
| | - Zarana S. Patel
- NASA Johnson Space Center, Houston, TX, United States
- KBR Inc., Houston, TX, United States
| | - Janice L. Huff
- NASA Langley Research Center, Hampton, VA, United States
| |
Collapse
|
7
|
Simmons P, Corley C, Allen AR. Fractionated Proton Irradiation Does Not Impair Hippocampal-Dependent Short-Term or Spatial Memory in Female Mice. TOXICS 2022; 10:toxics10090507. [PMID: 36136472 PMCID: PMC9503909 DOI: 10.3390/toxics10090507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/24/2022] [Accepted: 08/25/2022] [Indexed: 05/14/2023]
Abstract
The environment outside the Earth's protective magnetosphere is a much more threatening and complex space environment. The dominant causes for radiation exposure, solar particle events and galactic cosmic rays, contain high-energy protons. In space, astronauts need healthy and highly functioning cognitive abilities, of which the hippocampus plays a key role. Therefore, understanding the effects of 1H exposure on hippocampal-dependent cognition is vital for developing mitigative strategies and protective countermeasures for future missions. To investigate these effects, we subjected 6-month-old female CD1 mice to 0.75 Gy fractionated 1H (250 MeV) whole-body irradiation at the NASA Space Radiation Laboratory. The cognitive performance of the mice was tested 3 months after irradiation using Y-maze and Morris water maze tests. Both sham-irradiated and 1H-irradiated mice significantly preferred exploration of the novel arm compared to the familiar and start arms, indicating intact spatial and short-term memory. Both groups statistically spent more time in the target quadrant, indicating spatial memory retention. There were no significant differences in neurogenic and gliogenic cell counts after irradiation. In addition, proteomic analysis revealed no significant upregulation or downregulation of proteins related to behavior, neurological disease, or neural morphology. Our data suggests 1H exposure does not impair hippocampal-dependent spatial or short-term memory in female mice.
Collapse
Affiliation(s)
- Pilar Simmons
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, 4301 W Markham, Little Rock, AR 72205, USA
| | - Christa Corley
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, 4301 W Markham, Little Rock, AR 72205, USA
| | - Antiño R. Allen
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, 4301 W Markham, Little Rock, AR 72205, USA
- Correspondence: ; Tel.: +1-501-686-7553
| |
Collapse
|
8
|
Implication of Adult Hippocampal Neurogenesis in Alzheimer’s Disease and Potential Therapeutic Approaches. Cells 2022; 11:cells11020286. [PMID: 35053402 PMCID: PMC8773637 DOI: 10.3390/cells11020286] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 12/31/2022] Open
Abstract
Alzheimer’s disease is the most common neurodegenerative disease, affecting more than 6 million US citizens and representing the most prevalent cause for dementia. Neurogenesis has been repeatedly reported to be impaired in AD mouse models, but the reason for this impairment remains unclear. Several key factors play a crucial role in AD including Aβ accumulation, intracellular neurofibrillary tangles accumulation, and neuronal loss (specifically in the dentate gyrus of the hippocampus). Neurofibrillary tangles have been long associated with the neuronal loss in the dentate gyrus. Of note, Aβ accumulation plays an important role in the impairment of neurogenesis, but recent studies started to shed a light on the role of APP gene expression on the neurogenesis process. In this review, we will discuss the recent approaches to neurogenesis in Alzheimer disease and update the development of therapeutic methods.
Collapse
|
9
|
Dodson M, Anandhan A, Zhang DD, Madhavan L. An NRF2 Perspective on Stem Cells and Ageing. FRONTIERS IN AGING 2021; 2:690686. [PMID: 36213179 PMCID: PMC9536878 DOI: 10.3389/fragi.2021.690686] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 06/03/2021] [Indexed: 04/24/2023]
Abstract
Redox and metabolic mechanisms lie at the heart of stem cell survival and regenerative activity. NRF2 is a major transcriptional controller of cellular redox and metabolic homeostasis, which has also been implicated in ageing and lifespan regulation. However, NRF2's role in stem cells and their functioning with age is only just emerging. Here, focusing mainly on neural stem cells, which are core to adult brain plasticity and function, we review recent findings that identify NRF2 as a fundamental player in stem cell biology and ageing. We also discuss NRF2-based molecular programs that may govern stem cell state and function with age, and implications of this for age-related pathologies.
Collapse
Affiliation(s)
- Matthew Dodson
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, United States
| | - Annadurai Anandhan
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, United States
- Department of Neurology, University of Arizona, Tucson, AZ, United States
| | - Donna D. Zhang
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, United States
| | - Lalitha Madhavan
- Department of Neurology, University of Arizona, Tucson, AZ, United States
- Evelyn F. McKnight Brain Institute and Bio5 Institute, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
10
|
Huang Y, Mei X, Jiang W, Zhao H, Yan Z, Zhang H, Liu Y, Hu X, Zhang J, Peng W, Zhang J, Qi Q, Chen N. Mesenchymal Stem Cell-Conditioned Medium Protects Hippocampal Neurons From Radiation Damage by Suppressing Oxidative Stress and Apoptosis. Dose Response 2021; 19:1559325820984944. [PMID: 33716588 PMCID: PMC7923989 DOI: 10.1177/1559325820984944] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 12/06/2020] [Accepted: 12/07/2020] [Indexed: 11/15/2022] Open
Abstract
Objective To investigate the effects of mesenchymal stem cell-conditioned medium (MSC-CM) on radiation-induced oxidative stress, survival and apoptosis in hippocampal neurons. Methods The following groups were defined: Control, radiation treatment (RT), RT+MSC-CM, MSC-CM, RT + N-Acetylcysteine (RT+NAC), and RT + MSC-CM + PI3 K inhibitor (LY294002). A cell Counting Kit-8 (CCK-8) was used to measure cell proliferation. Apoptosis was examined by AnnexinV/PI flow cytometric analyses. Intracellular reactive oxygen species (ROS) were detected by DCFH-DA. Intracellular glutathione (GSH), malondialdehyde (MDA) content, and superoxide dismutase (SOD) activity were detected by colorimetric assays. Protein levels of γ-H2AX, PI3K-AKT, P53, cleaved caspase-3, Bax, and BCl-2 were analyzed by Western blotting. Results The proliferation of HT22 cells was significantly inhibited in the RT group, but was significantly preserved in the RT + MSC-CM group (P < 0.01). Apoptosis was significantly higher in the RT group than in the RT+ MSC-CM group (P < 0.01). MSC-CM decreased intracellular ROS and MDA content after irradiation (P < 0.01). GSH level and SOD activity were higher in the RT + MSC-CM group than in the RT group, as was MMP (P < 0.01). MSC-CM decreased expression of γ-H2AX, P53, Bax, and cleaved-caspase-3, but increased Bcl-2 expression (P < 0.01). Conclusion MSC-CM attenuated radiation-induced hippocampal neuron cell line damage by alleviating oxidative stress and suppressing apoptosis.
Collapse
Affiliation(s)
- Yue Huang
- North China University of Science and Technology, Tangshan, Hebei Province, China
| | - Xiaolong Mei
- Department of Orthopaedics, Tianjin Hospital, Tianjin, China
| | - Weishi Jiang
- North China University of Science and Technology, Tangshan, Hebei Province, China
| | - Hui Zhao
- Tianjin Key Laboratory of Food and Biotechnology, State Experimental and Training Centre of Food and Drug, School of Biotechnology and Food Science, Tianjin University of Commerce, Beichen, Tianjin, China
| | - Zhenyu Yan
- Department of Hematology, Affiliated Hospital of North China University of Science and Technology, Tangshan, Hebei Province, China
| | - Haixia Zhang
- Department of Hematology, Affiliated Hospital of North China University of Science and Technology, Tangshan, Hebei Province, China
| | - Ying Liu
- North China University of Science and Technology, Tangshan, Hebei Province, China
| | - Xia Hu
- North China University of Science and Technology, Tangshan, Hebei Province, China
| | - Jingyi Zhang
- North China University of Science and Technology, Tangshan, Hebei Province, China
| | - Wenshuo Peng
- North China University of Science and Technology, Tangshan, Hebei Province, China
| | - Jing Zhang
- The Third Central Hospital of Tianjin, Hedong District, Tianjin, China.,Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China.,Artificial Cell Engineering Technology Research Center, Tianjin, China.,Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Qingling Qi
- Department of Anesthesiology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Naiyao Chen
- Department of Hematology, Affiliated Hospital of North China University of Science and Technology, Tangshan, Hebei Province, China
| |
Collapse
|
11
|
Zhang B, Wang L, Zhan A, Wang M, Tian L, Guo W, Pan Y. Long-term exposure to a hypomagnetic field attenuates adult hippocampal neurogenesis and cognition. Nat Commun 2021; 12:1174. [PMID: 33608552 PMCID: PMC7896063 DOI: 10.1038/s41467-021-21468-x] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 01/19/2021] [Indexed: 12/14/2022] Open
Abstract
Adult hippocampal neurogenesis contributes to learning and memory, and is sensitive to a variety of environmental stimuli. Exposure to a hypomagnetic field (HMF) influences the cognitive processes of various animals, from insects to human beings. However, whether HMF exposure affect adult hippocampal neurogenesis and hippocampus-dependent cognitions is still an enigma. Here, we showed that male C57BL/6 J mice exposed to HMF by means of near elimination of the geomagnetic field (GMF) exhibit significant impairments of adult hippocampal neurogenesis and hippocampus-dependent learning, which is strongly correlated with a reduction in the content of reactive oxygen species (ROS). However, these deficits seen in HMF-exposed mice could be rescued either by elevating ROS levels through pharmacological inhibition of ROS removal or by returning them back to GMF. Therefore, our results suggest that GMF plays an important role in adult hippocampal neurogenesis through maintaining appropriate endogenous ROS levels.
Collapse
Affiliation(s)
- Bingfang Zhang
- Biogeomagnetism Group, Key Laboratory of Earth and Planetary Physics, Institute of Geology and Geophysics, Chinese Academy of Sciences, Beijing, China
- Innovation Academy for Earth Science, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Lei Wang
- University of Chinese Academy of Sciences, Beijing, China
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Aisheng Zhan
- Biogeomagnetism Group, Key Laboratory of Earth and Planetary Physics, Institute of Geology and Geophysics, Chinese Academy of Sciences, Beijing, China
- Innovation Academy for Earth Science, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Min Wang
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Lanxiang Tian
- Biogeomagnetism Group, Key Laboratory of Earth and Planetary Physics, Institute of Geology and Geophysics, Chinese Academy of Sciences, Beijing, China.
- Innovation Academy for Earth Science, Chinese Academy of Sciences, Beijing, China.
- The Paleomagnetism and Geochronology Laboratory, Institute of Geology and Geophysics, Chinese Academy of Sciences, Beijing, China.
| | - Weixiang Guo
- University of Chinese Academy of Sciences, Beijing, China.
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.
| | - Yongxin Pan
- Biogeomagnetism Group, Key Laboratory of Earth and Planetary Physics, Institute of Geology and Geophysics, Chinese Academy of Sciences, Beijing, China
- Innovation Academy for Earth Science, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- The Paleomagnetism and Geochronology Laboratory, Institute of Geology and Geophysics, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
12
|
Ferreira MKM, Aragão WAB, Bittencourt LO, Puty B, Dionizio A, Souza MPCD, Buzalaf MAR, de Oliveira EH, Crespo-Lopez ME, Lima RR. Fluoride exposure during pregnancy and lactation triggers oxidative stress and molecular changes in hippocampus of offspring rats. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 208:111437. [PMID: 33096359 DOI: 10.1016/j.ecoenv.2020.111437] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 09/24/2020] [Accepted: 09/28/2020] [Indexed: 05/28/2023]
Abstract
Long-term exposure to high concentrations of fluoride (F) can damage mineralized and soft tissues such as bones, liver, kidney, intestine, and nervous system of adult rats. The high permeability of the blood-brain barrier and placenta to F during pregnancy and lactation may be critical to neurological development. Therefore, this study aimed to investigate the effects of F exposure during pregnancy and lactation on molecular processes and oxidative biochemistry of offspring rats' hippocampus. Pregnant Wistar rats were randomly assigned into 3 groups in accordance with the drinking water received: G1 - deionized water (control); G2 - 10 mg/L of F and G3 - 50 mg/L of F. The exposure to fluoridated water began on the first day of pregnancy and lasted until the 21st day of breastfeeding (when the offspring rats were weaned). Blood plasma samples of the offspring rats were collected to determine F levels. Hippocampi samples were collected for oxidative biochemistry analyses through antioxidant capacity against peroxyl (ACAP), lipid peroxidation (LPO), and nitrite (NO2-) levels. Also, brain-derived neurotrophic factor (BDNF) gene expression (RT-qPCR) and proteomic profile analyses were performed. The results showed that exposure to both F concentrations during pregnancy and lactation increased the F bioavailability, triggered redox imbalance featured by a decrease of ACAP, increase of LPO and NO2- levels, BDNF overexpression and changes in the hippocampus proteome. These findings raise novel questions regarding potential repercussions on the hippocampus structure and functioning in the different cognitive domains.
Collapse
Affiliation(s)
- Maria Karolina Martins Ferreira
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Walessa Alana Bragança Aragão
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Leonardo Oliveira Bittencourt
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Bruna Puty
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Aline Dionizio
- Department of Biological Sciences, Bauru Dental School, University of São Paulo, Bauru, São Paulo, Brazil
| | | | | | | | - Maria Elena Crespo-Lopez
- Laboratory of Molecular Pharmacology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Rafael Rodrigues Lima
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil.
| |
Collapse
|
13
|
Davis CM, Allen AR, Bowles DE. Consequences of space radiation on the brain and cardiovascular system. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART C, TOXICOLOGY AND CARCINOGENESIS 2021; 39:180-218. [PMID: 33902387 DOI: 10.1080/26896583.2021.1891825] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Staying longer in outer space will inevitably increase the health risks of astronauts due to the exposures to galactic cosmic rays and solar particle events. Exposure may pose a significant hazard to space flight crews not only during the mission but also later, when slow-developing adverse effects could finally become apparent. The body of literature examining ground-based outcomes in response to high-energy charged-particle radiation suggests differential effects in response to different particles and energies. Numerous animal and cellular models have repeatedly demonstrated the negative effects of high-energy charged-particle on the brain and cognitive function. However, research on the role of space radiation in potentiating cardiovascular dysfunction is still in its early stages. This review summarizes the available data from studies using ground-based animal models to evaluate the response of the brain and heart to the high-energy charged particles of GCR and SPE, addresses potential sex differences in these effects, and aims to highlight gaps in the current literature for future study.
Collapse
Affiliation(s)
- Catherine M Davis
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences Bethesda, MD, USA
| | - Antiño R Allen
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Dawn E Bowles
- Division of Surgical Sciences, Department of Surgery, Duke University, Durham, NC, USA
| |
Collapse
|
14
|
Nicaise AM, Willis CM, Crocker SJ, Pluchino S. Stem Cells of the Aging Brain. Front Aging Neurosci 2020; 12:247. [PMID: 32848716 PMCID: PMC7426063 DOI: 10.3389/fnagi.2020.00247] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 07/20/2020] [Indexed: 12/12/2022] Open
Abstract
The adult central nervous system (CNS) contains resident stem cells within specific niches that maintain a self-renewal and proliferative capacity to generate new neurons, astrocytes, and oligodendrocytes throughout adulthood. Physiological aging is associated with a progressive loss of function and a decline in the self-renewal and regenerative capacities of CNS stem cells. Also, the biggest risk factor for neurodegenerative diseases is age, and current in vivo and in vitro models of neurodegenerative diseases rarely consider this. Therefore, combining both aging research and appropriate interrogation of animal disease models towards the understanding of the disease and age-related stem cell failure is imperative to the discovery of new therapies. This review article will highlight the main intrinsic and extrinsic regulators of neural stem cell (NSC) aging and discuss how these factors impact normal homeostatic functions within the adult brain. We will consider established in vivo animal and in vitro human disease model systems, and then discuss the current and future trajectories of novel senotherapeutics that target aging NSCs to ameliorate brain disease.
Collapse
Affiliation(s)
- Alexandra M Nicaise
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - Cory M Willis
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - Stephen J Crocker
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Stefano Pluchino
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
15
|
Terzi A, Suter DM. The role of NADPH oxidases in neuronal development. Free Radic Biol Med 2020; 154:33-47. [PMID: 32370993 DOI: 10.1016/j.freeradbiomed.2020.04.027] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 04/20/2020] [Accepted: 04/27/2020] [Indexed: 12/15/2022]
Abstract
Reactive oxygen species (ROS) are critical for maintaining cellular homeostasis and function when produced in physiological ranges. Important sources of cellular ROS include NADPH oxidases (Nox), which are evolutionary conserved multi-subunit transmembrane proteins. Nox-mediated ROS regulate variety of biological processes including hormone synthesis, calcium signaling, cell migration, and immunity. ROS participate in intracellular signaling by introducing post-translational modifications to proteins and thereby altering their functions. The central nervous system (CNS) expresses different Nox isoforms during both development and adulthood. Here, we review the role of Nox-mediated ROS during CNS development. Specifically, we focus on how individual Nox isoforms contribute to signaling in neural stem cell maintenance and neuronal differentiation, as well as neurite outgrowth and guidance. We also discuss how ROS regulates the organization and dynamics of the actin cytoskeleton in the neuronal growth cone. Finally, we review recent evidence that Nox-derived ROS modulate axonal regeneration upon nervous system injury.
Collapse
Affiliation(s)
- Aslihan Terzi
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA; Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, 47907, USA
| | - Daniel M Suter
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA; Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, 47907, USA; Bindley Bioscience Center, Purdue University, West Lafayette, IN, 47907, USA.
| |
Collapse
|
16
|
Mayer M, Arrizabalaga O, Ciba M, Schroeder IS, Ritter S, Thielemann C. Novel in vitro assay to investigate radiation induced changes in the functionality of human embryonic stem cell-derived neurospheres. Neurotoxicology 2020; 79:40-47. [PMID: 32320710 DOI: 10.1016/j.neuro.2020.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 03/31/2020] [Accepted: 04/15/2020] [Indexed: 10/24/2022]
Abstract
Ionizing radiation (IR) is increasingly used for diagnostics and therapy of severe brain diseases. However, IR also has adverse effects on the healthy brain tissue, particularly on the neuronal network. This is true for adults but even more pronounced in the developing brain of unborn and pediatric patients. Epidemiological studies on children receiving radiotherapy showed an increased risk for cognitive decline ranging from mild deficits in academic functioning to severe late effects in intellectual ability and language as a consequence of altered neuronal development and connectivity. To provide a comprehensive approach for the analysis of radiation-induced alterations in human neuronal functionality, we developed an in vitro assay by combining microelectrode array (MEA) analyses and human embryonic stem cell (hESC) derived three-dimensional neurospheres (NS). In our proof of principle study, we irradiated hESC with 1 Gy X-rays and let them spontaneously differentiate into neurons within NS. After the onset of neuronal activity, we recorded and analyzed the activity pattern of the developing neuronal networks. The network activity in NS derived from irradiated hESC was significantly reduced, whereas no differences in molecular endpoints such as cell proliferation and transcript or protein expression analyses were found. Thus, the combination of MEA analysis with a 3D model for neuronal functionality revealed radiation sequela that otherwise would not have been detected. We therefore strongly suggest combining traditional biomolecular methods with the new functional assay presented in this work to improve the risk assessment for IR-induced effects on the developing brain.
Collapse
Affiliation(s)
- Margot Mayer
- TH Aschaffenburg University of Applied Sciences, BioMEMS Lab, Aschaffenburg, Germany.
| | - Onetsine Arrizabalaga
- GSI Helmholtzzentrum für Schwerionenforschung, Biophysics Division, Darmstadt, Germany.
| | - Manuel Ciba
- TH Aschaffenburg University of Applied Sciences, BioMEMS Lab, Aschaffenburg, Germany.
| | - Insa S Schroeder
- GSI Helmholtzzentrum für Schwerionenforschung, Biophysics Division, Darmstadt, Germany.
| | - Sylvia Ritter
- GSI Helmholtzzentrum für Schwerionenforschung, Biophysics Division, Darmstadt, Germany.
| | - Christiane Thielemann
- TH Aschaffenburg University of Applied Sciences, BioMEMS Lab, Aschaffenburg, Germany.
| |
Collapse
|
17
|
Khacho M, Harris R, Slack RS. Mitochondria as central regulators of neural stem cell fate and cognitive function. Nat Rev Neurosci 2019; 20:34-48. [PMID: 30464208 DOI: 10.1038/s41583-018-0091-3] [Citation(s) in RCA: 254] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Emerging evidence now indicates that mitochondria are central regulators of neural stem cell (NSC) fate decisions and are crucial for both neurodevelopment and adult neurogenesis, which in turn contribute to cognitive processes in the mature brain. Inherited mutations and accumulated damage to mitochondria over the course of ageing serve as key factors underlying cognitive defects in neurodevelopmental disorders and neurodegenerative diseases, respectively. In this Review, we explore the recent findings that implicate mitochondria as crucial regulators of NSC function and cognition. In this respect, mitochondria may serve as targets for stem-cell-based therapies and interventions for cognitive defects.
Collapse
Affiliation(s)
- Mireille Khacho
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology (OISB), University of Ottawa, Ottawa, Ontario, Canada
| | - Richard Harris
- Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research Institute, Ottawa, Ontario, Canada
| | - Ruth S Slack
- Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research Institute, Ottawa, Ontario, Canada.
| |
Collapse
|
18
|
Yeo IJ, Park MH, Son DJ, Kim JY, Nam KT, Hyun BK, Kim SY, Jung MH, Song MJ, Chun HO, Lee TH, Han SB, Hong JT. PRDX6 Inhibits Neurogenesis through Downregulation of WDFY1-Mediated TLR4 Signal. Mol Neurobiol 2019; 56:3132-3144. [PMID: 30097850 PMCID: PMC6476867 DOI: 10.1007/s12035-018-1287-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Accepted: 07/30/2018] [Indexed: 12/22/2022]
Abstract
Impaired neurogenesis has been associated with several brain disorders, such as Alzheimer's disease (AD) and Parkinson's disease (PD). The role of peroxiredoxin 6 (PRDX6) in neurodegenerative diseases is very controversial. To demonstrate the role of PRDX6 in neurogenesis, we compared the neurogenesis ability of PRDX6-overexpressing transgenic (Tg) mice and wild-type mice and studied the involved molecular mechanisms. We showed that the neurogenesis of neural stem cells (NSCs) and the expression of the marker protein were lower in PRDX6 Tg-mice than in wild-type mice. To determine the factors involved in PRDX6-related neural stem cell impairment, we performed a microarray experiment. We showed that the expression of WDFY1 was dramatically decreased in PRDX6-Tg mice. Moreover, WDFY1 siRNA decreases the differentiation ability of primary neural stem cells. Interestingly, WDFY1 reportedly recruits the signaling adaptor TIR-domain-containing adapter-inducing interferon-β (TRIF) to toll-like receptors (TLRs); thus, we showed the relationship among TLRs, PRDX6, and WDFY1. We showed that TLR4 was dramatically reduced in PRDX6 Tg mice, and reduced TLR4 expression and neurogenesis was reversed by the introduction of WDFY1 plasmid in the neural stem cells from PRDX6 Tg mice. This study indicated that PRDX6 inhibits the neurogenesis of neural precursor cells through TLR4-dependent downregulation of WDFY1 and suggested that the inhibitory effect of PRDX6 on neurogenesis play a role in the development of neurodegenerative diseases in the PRDX6 overexpressing transgenic mice.
Collapse
Affiliation(s)
- In Jun Yeo
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Heungdeok-gu, Cheongju, Chungbuk, 361-951, Republic of Korea
| | - Mi Hee Park
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Heungdeok-gu, Cheongju, Chungbuk, 361-951, Republic of Korea
| | - Dong Ju Son
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Heungdeok-gu, Cheongju, Chungbuk, 361-951, Republic of Korea
| | - Ji Young Kim
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Heungdeok-gu, Cheongju, Chungbuk, 361-951, Republic of Korea
| | - Kyoung Tak Nam
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Heungdeok-gu, Cheongju, Chungbuk, 361-951, Republic of Korea
| | - Byung Kook Hyun
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Heungdeok-gu, Cheongju, Chungbuk, 361-951, Republic of Korea
| | - So Young Kim
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Heungdeok-gu, Cheongju, Chungbuk, 361-951, Republic of Korea
| | - Myung Hee Jung
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Heungdeok-gu, Cheongju, Chungbuk, 361-951, Republic of Korea
| | - Min Ji Song
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Heungdeok-gu, Cheongju, Chungbuk, 361-951, Republic of Korea
| | - Hyung Ok Chun
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Heungdeok-gu, Cheongju, Chungbuk, 361-951, Republic of Korea
| | - Tae Hyung Lee
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Heungdeok-gu, Cheongju, Chungbuk, 361-951, Republic of Korea
| | - Sang-Bae Han
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Heungdeok-gu, Cheongju, Chungbuk, 361-951, Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Heungdeok-gu, Cheongju, Chungbuk, 361-951, Republic of Korea.
| |
Collapse
|
19
|
Mineyeva OA, Bezriadnov DV, Kedrov AV, Lazutkin AA, Anokhin KV, Enikolopov GN. Radiation Induces Distinct Changes in Defined Subpopulations of Neural Stem and Progenitor Cells in the Adult Hippocampus. Front Neurosci 2019; 12:1013. [PMID: 30686979 PMCID: PMC6333747 DOI: 10.3389/fnins.2018.01013] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Accepted: 12/17/2018] [Indexed: 11/13/2022] Open
Abstract
While irradiation can effectively treat brain tumors, this therapy also causes cognitive impairments, some of which may stem from the disruption of hippocampal neurogenesis. To study how radiation affects neurogenesis, we combine phenotyping of subpopulations of hippocampal neural stem and progenitor cells with double- and triple S-phase labeling paradigms. Using this approach, we reveal new features of division, survival, and differentiation of neural stem and progenitor cells after exposure to gamma radiation. We show that dividing neural stem cells, while susceptible to damage induced by gamma rays, are less vulnerable than their rapidly amplifying progeny. We also show that dividing stem and progenitor cells that survive irradiation are suppressed in their ability to replicate 0.5–1 day after the radiation exposure. Suppression of division is also observed for cells that entered the cell cycle after irradiation or were not in the S phase at the time of exposure. Determining the longer term effects of irradiation, we found that 2 months after exposure, radiation-induced suppression of division is partially relieved for both stem and progenitor cells, without evidence for compensatory symmetric divisions as a means to restore the normal level of neurogenesis. By that time, most mature young neurons, born 2–4 weeks after the irradiation, still bear the consequences of radiation exposure, unlike younger neurons undergoing early stages of differentiation without overt signs of deficient maturation. Later, 6 months after an exposure to 5 Gy, cell proliferation and neurogenesis are further impaired, though neural stem cells are still available in the niche, and their pool is preserved. Our results indicate that various subpopulations of stem and progenitor cells in the adult hippocampus have different susceptibility to gamma radiation, and that neurogenesis, even after a temporary restoration, is impaired in the long term after exposure to gamma rays. Our study provides a framework for investigating critical issues of neural stem cell maintenance, aging, interaction with their microenvironment, and post-irradiation therapy.
Collapse
Affiliation(s)
- Olga A Mineyeva
- Moscow Institute of Physics and Technology, Dolgoprudny, Russia.,P. K. Anokhin Research Institute of Normal Physiology, Moscow, Russia.,National Research Center "Kurchatov Institute," Moscow, Russia
| | - Dmitri V Bezriadnov
- Moscow Institute of Physics and Technology, Dolgoprudny, Russia.,P. K. Anokhin Research Institute of Normal Physiology, Moscow, Russia
| | - Alexander V Kedrov
- Moscow Institute of Physics and Technology, Dolgoprudny, Russia.,P. K. Anokhin Research Institute of Normal Physiology, Moscow, Russia
| | - Alexander A Lazutkin
- Moscow Institute of Physics and Technology, Dolgoprudny, Russia.,P. K. Anokhin Research Institute of Normal Physiology, Moscow, Russia.,N.N. Burdenko Neurosurgery Institute, Moscow, Russia
| | - Konstantin V Anokhin
- Moscow Institute of Physics and Technology, Dolgoprudny, Russia.,P. K. Anokhin Research Institute of Normal Physiology, Moscow, Russia.,National Research Center "Kurchatov Institute," Moscow, Russia
| | - Grigori N Enikolopov
- Moscow Institute of Physics and Technology, Dolgoprudny, Russia.,Center for Developmental Genetics, Department of Anesthesiology, Stony Brook University, Stony Brook, NY, United States
| |
Collapse
|
20
|
Feng X, Liu S, Chen D, Rosi S, Gupta N. Rescue of cognitive function following fractionated brain irradiation in a novel preclinical glioma model. eLife 2018; 7:e38865. [PMID: 30421720 PMCID: PMC6234025 DOI: 10.7554/elife.38865] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 10/24/2018] [Indexed: 11/13/2022] Open
Abstract
More than half of long-term brain tumor survivors develop irreversible cognitive decline that severely affect their quality of life. However, there is no pre-clinical model that allows long-term assessment of cognition, and there is no treatment which ameliorates cognitive deficits in patients. Here, we report a novel glioma mouse model that offers manageable tumor growth and reliable assessment of cognitive functions in a post-treatment manner. Using this model, we found that fractionated whole-brain irradiation (fWBI), but not tumor growth, results in memory deficits. Transient inhibition of CSF-1R during fWBI prolongs survival of glioma-bearing mice and fully prevents fWBI-induced memory deficits. This result suggests that CSF-1R inhibition during radiotherapy can be explored as an approach to improve both survival and cognitive outcomes in patients who will receive fWBI. Taken together, the current study provides a proof of concept of a powerful tool to study radiation-induced cognitive deficits in glioma-bearing animals.
Collapse
Affiliation(s)
- Xi Feng
- Brain and Spinal Injury CenterUniversity of California San FranciscoSan FranciscoUnited States
- Department of Physical Therapy and Rehabilitation ScienceUniversity of California San FranciscoSan FranciscoUnited States
| | - Sharon Liu
- Department of Neurological SurgeryUniversity of California San FranciscoSan FranciscoUnited States
- Brain Tumor Research CenterUniversity of California San FranciscoSan FranciscoUnited States
| | - David Chen
- Department of Neurological SurgeryUniversity of California San FranciscoSan FranciscoUnited States
| | - Susanna Rosi
- Brain and Spinal Injury CenterUniversity of California San FranciscoSan FranciscoUnited States
- Department of Physical Therapy and Rehabilitation ScienceUniversity of California San FranciscoSan FranciscoUnited States
- Department of Neurological SurgeryUniversity of California San FranciscoSan FranciscoUnited States
- Weill Institute for NeuroscienceUniversity of California San FranciscoSan FranciscoUnited States
- Kavli Institute of Fundamental NeuroscienceUniversity of California San FranciscoSan FranciscoUnited States
| | - Nalin Gupta
- Department of Neurological SurgeryUniversity of California San FranciscoSan FranciscoUnited States
- Brain Tumor Research CenterUniversity of California San FranciscoSan FranciscoUnited States
| |
Collapse
|
21
|
Cho HJ, Lee WH, Hwang OMH, Sonntag WE, Lee YW. Role of NADPH oxidase in radiation-induced pro-oxidative and pro-inflammatory pathways in mouse brain. Int J Radiat Biol 2017; 93:1257-1266. [PMID: 28880721 DOI: 10.1080/09553002.2017.1377360] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE The present study was designed to investigate our hypothesis that NADPH oxidase plays a role in radiation-induced pro-oxidative and pro-inflammatory environments in the brain. MATERIALS AND METHODS C57BL/6 mice received either fractionated whole brain irradiation or sham-irradiation. The mRNA expression levels of pro-inflammatory mediators, such as TNF-α and MCP-1, were determined by quantitative real-time RT-PCR. The protein expression levels of TNF-α, MCP-1, NOX-2 and Iba1 were detected by immunofluorescence staining. The levels of ROS were visualized by in situ DHE fluorescence staining. RESULTS A significant up-regulation of mRNA and protein expression levels of TNF-α and MCP-1 was observed in irradiated mouse brains. Additionally, immunofluorescence staining of Iba1 showed a marked increase of microglial activation in mouse brain after irradiation. Moreover, in situ DHE fluorescence staining revealed that fractionated whole brain irradiation significantly increased production of ROS. Furthermore, a significant increase in immunoreactivity of NOX-2 was detected in mouse brain after irradiation. On the contrary, an enhanced ROS generation in mouse brain after irradiation was markedly attenuated in the presence of NOX inhibitors or NOX-2 neutralizing antibody. CONCLUSIONS These results suggest that NOX-2 may play a role in fractionated whole brain irradiation-induced pro-oxidative and pro-inflammatory pathways in mouse brain.
Collapse
Affiliation(s)
- Hyung Joon Cho
- a Department of Biochemistry and Molecular Biology , University of Miami Miller School of Medicine , Miami , FL , USA
| | - Won Hee Lee
- b Stanford Cardiovascular Institute , Stanford University , Stanford , CA , USA
| | - Olivia Min Ha Hwang
- c Department of Biomedical Engineering and Mechanics , Virginia Tech , Blacksburg , VA , USA
| | - William E Sonntag
- d Department of Geriatric Medicine , University of Oklahoma Health Sciences Center , Oklahoma City , OK , USA
| | - Yong Woo Lee
- c Department of Biomedical Engineering and Mechanics , Virginia Tech , Blacksburg , VA , USA
| |
Collapse
|
22
|
Hofer T, Duale N, Muusse M, Eide DM, Dahl H, Boix F, Andersen JM, Olsen AK, Myhre O. Restoration of Cognitive Performance in Mice Carrying a Deficient Allele of 8-Oxoguanine DNA Glycosylase by X-ray Irradiation. Neurotox Res 2017; 33:824-836. [DOI: 10.1007/s12640-017-9833-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 10/13/2017] [Accepted: 10/18/2017] [Indexed: 12/13/2022]
|
23
|
Durak MA, Parlakpinar H, Polat A, Vardi N, Ekici K, Ucar M, Ozhan O, Yildiz A, Pasahan R. Protective and therapeutic effects of molsidomine on radiation induced neural injury in rats. Biotech Histochem 2017; 92:68-77. [DOI: 10.1080/10520295.2016.1271454] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Affiliation(s)
| | | | | | | | | | - M Ucar
- Anesthesiology, Medical School, Inonu University, Malatya, Turkey
| | | | | | | |
Collapse
|
24
|
Kukoamine A Prevents Radiation-Induced Neuroinflammation and Preserves Hippocampal Neurogenesis in Rats by Inhibiting Activation of NF-κB and AP-1. Neurotox Res 2016; 31:259-268. [DOI: 10.1007/s12640-016-9679-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 10/16/2016] [Accepted: 10/18/2016] [Indexed: 12/18/2022]
|
25
|
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
|
26
|
Villasana LE, Weber S, Akinyeke T, Raber J. Genotype differences in anxiety and fear learning and memory of WT and ApoE4 mice associated with enhanced generation of hippocampal reactive oxygen species. J Neurochem 2016; 138:896-908. [PMID: 27412623 DOI: 10.1111/jnc.13737] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 06/28/2016] [Accepted: 06/29/2016] [Indexed: 12/14/2022]
Abstract
Apolipoprotein E (apoE), involved in cholesterol and lipid metabolism, also influences cognitive function and injury repair. In humans, apoE is expressed in three isoforms. E4 is a risk factor for age-related cognitive decline and Alzheimer's disease, particularly in women. E4 might also be a risk factor for developing behavioral and cognitive changes following (56) Fe irradiation, a component of the space environment astronauts are exposed to during missions. These changes might be related to enhanced generation of reactive oxygen species (ROS). In this study, we compared the behavioral and cognitive performance of sham-irradiated and irradiated wild-type (WT) mice and mice expressing the human E3 or E4 isoforms, and assessed the generation of ROS in hippocampal slices from these mice. E4 mice had greater anxiety-like and conditioned fear behaviors than WT mice, and these genotype differences were associated with greater levels of ROS in E4 than WT mice. The greater generation of ROS in the hippocampus of E4 than WT mice might contribute to their higher anxiety levels and enhanced fear conditioning. In E4, but not WT, mice, phorbol-12-myristate-13-acetate-treated hippocampal slices showed more dihydroxy ethidium oxidation in sham-irradiated than irradiated mice and hippocampal heme oxygenase-1 levels were higher in irradiated than sham-irradiated E4 mice. Mice with apolipoprotein E4 (E4), a risk factor for Alzheimer's disease, have greater anxiety-like and conditioned fear behaviors than wild-type (WT) mice. Generation of reactive oxygen species (ROS, in red) 3 months following (56) Fe irradiation, a component of the space environment astronauts are exposed to, is more pronounced in the hippocampus of E4 than WT mice. In E4, but not WT, mice, hippocampal levels of the oxidative stress-relevant marker heme oxygenase-1 are higher in irradiated than sham-irradiated E4 mice.
Collapse
Affiliation(s)
- Laura E Villasana
- Division of Neuroscience, Department of Behavioral Neuroscience, ONPRC, Oregon Health & Science University, Portland, Oregon, USA
| | - Sydney Weber
- Division of Neuroscience, Department of Behavioral Neuroscience, ONPRC, Oregon Health & Science University, Portland, Oregon, USA
| | - Tunde Akinyeke
- Division of Neuroscience, Department of Behavioral Neuroscience, ONPRC, Oregon Health & Science University, Portland, Oregon, USA
| | - Jacob Raber
- Division of Neuroscience, Department of Behavioral Neuroscience, ONPRC, Oregon Health & Science University, Portland, Oregon, USA. .,Division of Neuroscience, Departments of Neurology and Radiation Medicine, ONPRC, Oregon Health & Science University, Portland, Oregon, USA.
| |
Collapse
|
27
|
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
|
28
|
Yuan TF, Gu S, Shan C, Marchado S, Arias-Carrión O. Oxidative Stress and Adult Neurogenesis. Stem Cell Rev Rep 2016; 11:706-9. [PMID: 26100529 DOI: 10.1007/s12015-015-9603-y] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
There is a growing evidence that adult neurogenesis is critical for brain function. The reactive oxygen species (ROS) is accumulated during adult neurogenesis as a physiological mechanism; while ROS overload impairs adult neurogenesis during ageing, neuroinflammation and neurodegeneration. Here we propose that targeting oxidative stress provides a novel way to regulate adult neurogenesis and manage different brain diseases.
Collapse
Affiliation(s)
- Ti-Fei Yuan
- School of Psychology, Nanjing Normal University, Nanjing, China,
| | | | | | | | | |
Collapse
|
29
|
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
|
30
|
Zhang Y, Cheng Z, Wang C, Ma H, Meng W, Zhao Q. Neuroprotective Effects of Kukoamine a against Radiation-induced Rat Brain Injury through Inhibition of Oxidative Stress and Neuronal Apoptosis. Neurochem Res 2016; 41:2549-2558. [DOI: 10.1007/s11064-016-1967-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 05/24/2016] [Accepted: 05/25/2016] [Indexed: 11/29/2022]
|
31
|
Chen H, Goodus MT, de Toledo SM, Azzam EI, Levison SW, Souayah N. Ionizing Radiation Perturbs Cell Cycle Progression of Neural Precursors in the Subventricular Zone Without Affecting Their Long-Term Self-Renewal. ASN Neuro 2015; 7:7/3/1759091415578026. [PMID: 26056396 PMCID: PMC4461572 DOI: 10.1177/1759091415578026] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Damage to normal human brain cells from exposure to ionizing radiation may occur during the course of radiotherapy or from accidental exposure. Delayed effects may complicate the immediate effects resulting in neurodegeneration and cognitive decline. We examined cellular and molecular changes associated with exposure of neural stem/progenitor cells (NSPs) to 137Cs γ-ray doses in the range of 0 to 8 Gy. Subventricular zone NSPs isolated from newborn mouse pups were analyzed for proliferation, self-renewal, and differentiation, shortly after irradiation. Strikingly, there was no apparent increase in the fraction of dying cells after irradiation, and the number of single cells that formed neurospheres showed no significant change from control. Upon differentiation, irradiated neural precursors did not differ in their ability to generate neurons, astrocytes, and oligodendrocytes. By contrast, progression of NSPs through the cell cycle decreased dramatically after exposure to 8 Gy (p < .001). Mice at postnatal day 10 were exposed to 8 Gy of γ rays delivered to the whole body and NSPs of the subventricular zone were analyzed using a four-color flow cytometry panel combined with ethynyl deoxyuridine incorporation. Similar flow cytometric analyses were performed on NSPs cultured as neurospheres. These studies revealed that neither the percentage of neural stem cells nor their proliferation was affected. By contrast, γ-irradiation decreased the proliferation of two classes of multipotent cells and increased the proliferation of a specific glial-restricted precursor. Altogether, these results support the conclusion that primitive neural precursors are radioresistant, but their proliferation is slowed down as a consequence of γ-ray exposure.
Collapse
Affiliation(s)
- Hongxin Chen
- Department of Neurology and Neurosciences, Rutgers University-New Jersey Medical School, Newark, NJ, USA
| | - Matthew T Goodus
- Department of Neurology and Neurosciences, Rutgers University-New Jersey Medical School, Newark, NJ, USA
| | - Sonia M de Toledo
- Department of Radiology, Rutgers University-New Jersey Medical School, Newark, NJ, USA
| | - Edouard I Azzam
- Department of Radiology, Rutgers University-New Jersey Medical School, Newark, NJ, USA
| | - Steven W Levison
- Department of Neurology and Neurosciences, Rutgers University-New Jersey Medical School, Newark, NJ, USA
| | - Nizar Souayah
- Department of Neurology and Neurosciences, Rutgers University-New Jersey Medical School, Newark, NJ, USA
| |
Collapse
|
32
|
Huang TT, Leu D, Zou Y. Oxidative stress and redox regulation on hippocampal-dependent cognitive functions. Arch Biochem Biophys 2015; 576:2-7. [PMID: 25797440 DOI: 10.1016/j.abb.2015.03.014] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 03/12/2015] [Accepted: 03/13/2015] [Indexed: 12/17/2022]
Abstract
Hippocampal-dependent cognitive functions rely on production of new neurons and maintenance of dendritic structures to provide the synaptic plasticity needed for learning and formation of new memories. Hippocampal formation is exquisitely sensitive to patho-physiological changes, and reduced antioxidant capacity and exposure to low dose irradiation can significantly impede hippocampal-dependent functions of learning and memory by reducing the production of new neurons and alter dendritic structures in the hippocampus. Although the mechanism leading to impaired cognitive functions is complex, persistent oxidative stress likely plays an important role in the SOD-deficient and radiation-exposed hippocampal environment. Aging is associated with increased production of pro-oxidants and accumulation of oxidative end products. Similar to the hippocampal defects observed in SOD-deficient mice and mice exposed to low dose irradiation, reduced capacity in learning and memory, diminishing hippocampal neurogenesis, and altered dendritic network are universal in the aging brains. Given the similarities in cellular and structural changes in the aged, SOD-deficient, and radiation-exposed hippocampal environment and the corresponding changes in cognitive decline, understanding the shared underlying mechanism will provide more flexible and efficient use of SOD deficiency or irradiation to model age-related changes in cognitive functions and identify potential therapeutic or intervention methods.
Collapse
Affiliation(s)
- Ting-Ting Huang
- Geriatric Research, Education, and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA, USA; Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.
| | - David Leu
- Geriatric Research, Education, and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA, USA; Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Yani Zou
- Geriatric Research, Education, and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA, USA; Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
33
|
Kazim SF, Cardenas-Aguayo MDC, Arif M, Blanchard J, Fayyaz F, Grundke-Iqbal I, Iqbal K. Sera from children with autism induce autistic features which can be rescued with a CNTF small peptide mimetic in rats. PLoS One 2015; 10:e0118627. [PMID: 25769033 PMCID: PMC4359103 DOI: 10.1371/journal.pone.0118627] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 01/21/2015] [Indexed: 12/29/2022] Open
Abstract
Autism is a neurodevelopmental disorder characterized clinically by impairments in social interaction and verbal and non-verbal communication skills as well as restricted interests and repetitive behavior. It has been hypothesized that altered brain environment including an imbalance in neurotrophic support during early development contributes to the pathophysiology of autism. Here we report that sera from children with autism which exhibited abnormal levels of various neurotrophic factors induced cell death and oxidative stress in mouse primary cultured cortical neurons. The effects of sera from autistic children were rescued by pre-treatment with a ciliary neurotrophic factor (CNTF) small peptide mimetic, Peptide 6 (P6), which was previously shown to exert its neuroprotective effect by modulating CNTF/JAK/STAT pathway and LIF signaling and by enhancing brain derived neurotrophic factor (BDNF) expression. Similar neurotoxic effects and neuroinflammation were observed in young Wistar rats injected intracerebroventricularly with autism sera within hours after birth. The autism sera injected rats demonstrated developmental delay and deficits in social communication, interaction, and novelty. Both the neurobiological changes and the behavioral autistic phenotype were ameliorated by P6 treatment. These findings implicate the involvement of neurotrophic imbalance during early brain development in the pathophysiology of autism and a proof of principle of P6 as a potential therapeutic strategy for autism.
Collapse
Affiliation(s)
- Syed Faraz Kazim
- Inge Grundke-Iqbal Research Floor, Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities (NYSIBR), Staten Island, New York, United States of America
- Neural and Behavioral Science Graduate Program, State University of New York (SUNY) Downstate Medical Center, Brooklyn, New York, United States of America
- SUNY Downstate/NYSIBR Center for Developmental Neuroscience (CDN), Staten Island, New York, United States of America
| | - Maria del Carmen Cardenas-Aguayo
- Inge Grundke-Iqbal Research Floor, Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities (NYSIBR), Staten Island, New York, United States of America
| | - Mohammad Arif
- Inge Grundke-Iqbal Research Floor, Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities (NYSIBR), Staten Island, New York, United States of America
| | - Julie Blanchard
- Inge Grundke-Iqbal Research Floor, Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities (NYSIBR), Staten Island, New York, United States of America
| | - Fatima Fayyaz
- Inge Grundke-Iqbal Research Floor, Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities (NYSIBR), Staten Island, New York, United States of America
| | - Inge Grundke-Iqbal
- Inge Grundke-Iqbal Research Floor, Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities (NYSIBR), Staten Island, New York, United States of America
| | - Khalid Iqbal
- Inge Grundke-Iqbal Research Floor, Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities (NYSIBR), Staten Island, New York, United States of America
- SUNY Downstate/NYSIBR Center for Developmental Neuroscience (CDN), Staten Island, New York, United States of America
- * E-mail:
| |
Collapse
|
34
|
Sokolova IV, Schneider CJ, Bezaire M, Soltesz I, Vlkolinsky R, Nelson GA. Proton Radiation Alters Intrinsic and Synaptic Properties of CA1 Pyramidal Neurons of the Mouse Hippocampus. Radiat Res 2015; 183:208-18. [DOI: 10.1667/rr13785.1] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Irina V. Sokolova
- Department of Basic Sciences, Division of Radiation Research, School of Medicine, Loma Linda University, Loma Linda, California
| | - Calvin J. Schneider
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, California
| | - Marianne Bezaire
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, California
| | - Ivan Soltesz
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, California
| | - Roman Vlkolinsky
- Department of Basic Sciences, Division of Radiation Research, School of Medicine, Loma Linda University, Loma Linda, California
| | - Gregory A. Nelson
- Department of Basic Sciences, Division of Radiation Research, School of Medicine, Loma Linda University, Loma Linda, California
| |
Collapse
|
35
|
Parihar VK, Allen BD, Tran KK, Chmielewski NN, Craver BM, Martirosian V, Morganti JM, Rosi S, Vlkolinsky R, Acharya MM, Nelson GA, Allen AR, Limoli CL. Targeted overexpression of mitochondrial catalase prevents radiation-induced cognitive dysfunction. Antioxid Redox Signal 2015; 22:78-91. [PMID: 24949841 PMCID: PMC4270160 DOI: 10.1089/ars.2014.5929] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
AIMS Radiation-induced disruption of mitochondrial function can elevate oxidative stress and contribute to the metabolic perturbations believed to compromise the functionality of the central nervous system. To clarify the role of mitochondrial oxidative stress in mediating the adverse effects of radiation in the brain, we analyzed transgenic (mitochondrial catalase [MCAT]) mice that overexpress human catalase localized to the mitochondria. RESULTS Compared with wild-type (WT) controls, overexpression of the MCAT transgene significantly decreased cognitive dysfunction after proton irradiation. Significant improvements in behavioral performance found on novel object recognition and object recognition in place tasks were associated with a preservation of neuronal morphology. While the architecture of hippocampal CA1 neurons was significantly compromised in irradiated WT mice, the same neurons in MCAT mice did not exhibit extensive and significant radiation-induced reductions in dendritic complexity. Irradiated neurons from MCAT mice maintained dendritic branching and length compared with WT mice. Protected neuronal morphology in irradiated MCAT mice was also associated with a stabilization of radiation-induced variations in long-term potentiation. Stabilized synaptic activity in MCAT mice coincided with an altered composition of the synaptic AMPA receptor subunits GluR1/2. INNOVATION Our findings provide the first evidence that neurocognitive sequelae associated with radiation exposure can be reduced by overexpression of MCAT, operating through a mechanism involving the preservation of neuronal morphology. CONCLUSION Our article documents the neuroprotective properties of reducing mitochondrial reactive oxygen species through the targeted overexpression of catalase and how this ameliorates the adverse effects of proton irradiation in the brain.
Collapse
Affiliation(s)
- Vipan K. Parihar
- Department of Radiation Oncology, University of California, Irvine, Irvine, California
| | - Barrett D. Allen
- Department of Radiation Oncology, University of California, Irvine, Irvine, California
| | - Katherine K. Tran
- Department of Radiation Oncology, University of California, Irvine, Irvine, California
| | - Nicole N. Chmielewski
- Department of Radiation Oncology, University of California, Irvine, Irvine, California
| | - Brianna M. Craver
- Department of Radiation Oncology, University of California, Irvine, Irvine, California
| | - Vahan Martirosian
- Department of Radiation Oncology, University of California, Irvine, Irvine, California
| | - Josh M. Morganti
- Departments of Physical Therapy Rehabilitation Science and Neurological Surgery, University of California, San Francisco, San Francisco, California
| | - Susanna Rosi
- Departments of Physical Therapy Rehabilitation Science and Neurological Surgery, University of California, San Francisco, San Francisco, California
- Brain and Spinal Injury Center, University of California, San Francisco, San Francisco, California
| | - Roman Vlkolinsky
- Departments of Radiation Medicine and Basic Sciences, Loma Linda University, Loma Linda, California
| | - Munjal M. Acharya
- Department of Radiation Oncology, University of California, Irvine, Irvine, California
| | - Gregory A. Nelson
- Departments of Radiation Medicine and Basic Sciences, Loma Linda University, Loma Linda, California
| | - Antiño R. Allen
- Division of Radiation Health, University of Arkansas Medical School, Little Rock, Arkansas
| | - Charles L. Limoli
- Department of Radiation Oncology, University of California, Irvine, Irvine, California
| |
Collapse
|
36
|
Zhang L, Li K, Sun R, Zhang Y, Ji J, Huang P, Yang H, Tian Y. Minocycline ameliorates cognitive impairment induced by whole-brain irradiation: an animal study. Radiat Oncol 2014; 9:281. [PMID: 25498371 PMCID: PMC4271325 DOI: 10.1186/s13014-014-0281-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 11/28/2014] [Indexed: 12/11/2022] Open
Abstract
Background It has been long recognized that cranial irradiation used for the treatment of primary and metastatic brain tumor often causes neurological side-effects such as intellectual impairment, memory loss and dementia, especially in children patients. Our previous study has demonstrated that whole-brain irradiation (WBI) can cause cognitive decline in rats. Minocycline is an antibiotic that has shown neuroprotective properties in a variety of experimental models of neurological diseases. However, whether minocycline can ameliorate cognitive impairment induced by ionizing radiation (IR) has not been tested. Thus this study aimed to demonstrate the potential implication of minocycline in the treatment of WBI-induced cognitive deficits by using a rat model. Methods Sprague Dawley rats were cranial irradiated with electron beams delivered by a linear accelerator with a single dose of 20 Gy. Minocycline was administered via oral gavages directly into the stomach before and after irradiation. The open field test was used to assess the anxiety level of rats. The Morris water maze (MWM) was used to assess the spatial learning and memory of rats. The level of apoptosis in hippocampal neurons was measured using immunohistochemistry for caspase-3 and relative markers for mature neurons (NeuN) or for newborn neurons (Doublecortin (DCX)). Neurogenesis was determined by BrdU incorporation method. Results Neither WBI nor minocycline affected the locomotor activity and anxiety level of rats. However, compared with the sham-irradiated controls, WBI caused a significant loss of learning and memory manifest as longer latency to reach the hidden platform in the MWM task. Minocycline intervention significantly improved the memory retention of irradiated rats. Although minocycline did not rescue neurogenesis deficit caused by WBI 2 months post-IR, it did significantly decreased WBI-induced apoptosis in the DCX positive neurons, thereby resulting in less newborn neuron depletion 12 h after irradiation. Conclusions Minocycline significantly inhibits WBI-induced neuron apoptosis, leading to less newborn neurons loss shortly after irradiation. In the long run, minocycline improves the cognitive performance of rats post WBI. The results indicate a potential clinical implication of minocycline as an effective adjunct in radiotherapy for brain tumor patients.
Collapse
Affiliation(s)
- Liyuan Zhang
- Department of Radiotherapy and Oncology, Second Affiliated Hospital, Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu Province, 215004, PR China. .,Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Second Affiliated Hospital, Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu Province, 215004, PR China. .,Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, Jiangsu Province, 215123, PR China.
| | - Kun Li
- Department of Radiotherapy and Oncology, Second Affiliated Hospital, Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu Province, 215004, PR China. .,Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Second Affiliated Hospital, Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu Province, 215004, PR China. .,Department of Medical Oncology, Affiliated Hospital of Taishan Medical University, Taian, Shandong Province, 271000, PR China.
| | - Rui Sun
- Department of Radiotherapy and Oncology, Second Affiliated Hospital, Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu Province, 215004, PR China. .,Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Second Affiliated Hospital, Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu Province, 215004, PR China.
| | - Yuan Zhang
- Department of Radiotherapy and Oncology, Second Affiliated Hospital, Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu Province, 215004, PR China. .,Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Second Affiliated Hospital, Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu Province, 215004, PR China.
| | - JianFeng Ji
- Department of Radiotherapy and Oncology, Second Affiliated Hospital, Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu Province, 215004, PR China. .,Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Second Affiliated Hospital, Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu Province, 215004, PR China.
| | - Peigeng Huang
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, 100 Blossom Street, Boston, MA, 02114, USA.
| | - Hongying Yang
- Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, Jiangsu Province, 215123, PR China. .,School of Radiation Medicine and Protection, Medical College of Soochow University/School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, 199 Renai Road, Suzhou Industrial Park, Suzhou, Jiangsu Province, 215123, PR China.
| | - Ye Tian
- Department of Radiotherapy and Oncology, Second Affiliated Hospital, Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu Province, 215004, PR China. .,Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Second Affiliated Hospital, Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu Province, 215004, PR China. .,Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, Jiangsu Province, 215123, PR China.
| |
Collapse
|
37
|
Marty VN, Vlkolinsky R, Minassian N, Cohen T, Nelson GA, Spigelman I. Radiation-Induced Alterations in Synaptic Neurotransmission of Dentate Granule Cells Depend on the Dose and Species of Charged Particles. Radiat Res 2014; 182:653-65. [DOI: 10.1667/rr13647.1] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
38
|
Allen AR, Eilertson K, Sharma S, Baure J, Allen B, Leu D, Rosi S, Raber J, Huang TT, Fike JR. Delayed administration of alpha-difluoromethylornithine prevents hippocampus-dependent cognitive impairment after single and combined injury in mice. Radiat Res 2014; 182:489-98. [PMID: 25375198 PMCID: PMC4282164 DOI: 10.1667/rr13753.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Radiation exposure due to radiological terrorism and military circumstances are a continuing threat for the civilian population. In an uncontrolled radiation event, it is likely that there will be other types of injury involved, including trauma. While radiation combined injury is recognized as an area of great significance, overall there is a paucity of information regarding the mechanisms underlying the interactions between irradiation and other forms of injury, or what countermeasures might be effective in ameliorating such changes. The objective of this study was to determine if difluoromethylornithine (DFMO) could reduce the adverse effects of single or combined injury if administered beginning 24 h after exposure. Eight-week-old C57BL/J6 young-adult male mice received whole-body cesium-137 ((137)Cs) irradiation with 4 Gy. Immediately after irradiation, unilateral traumatic brain injury was induced using a controlled cortical impact system. Forty-four days postirradiation, animals were tested for hippocampus-dependent cognitive performance in the Morris water maze. After cognitive testing, animals were euthanized and their brains snap frozen for immunohistochemical assessment of neuroinflammation (activated microglia) and neurogenesis in the hippocampal dentate gyrus. Our data show that single and combined injuries induced variable degrees of hippocampus-dependent cognitive dysfunction, and when given 24 h post trauma, DFMO treatment ameliorated those effects. Cellular changes including neurogenesis and numbers of activated microglia were generally not associated with the cognitive changes. Further analyses also revealed that DFMO increased hippocampal protein levels of the antioxidants thioredoxin 1 and peroxiredoxin 3 compared to vehicle treated animals. While the mechanisms responsible for the improvement in cognition after DFMO treatment are not yet clear, these results constitute a basis for further development of DFMO as a countermeasure for ameliorating the of risks for cognitive dysfunction in individuals subjected to trauma and radiation combined injury.
Collapse
Affiliation(s)
- Antiño R. Allen
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
| | - Kirsten Eilertson
- Bioinformatics Core, Gladstone Institutes San Francisco, California 94158
| | - Sourabh Sharma
- Brain and Spinal Injury Center, Department of Neurological Surgery, University of California, San Francisco, California 94110
| | - Jennifer Baure
- Brain and Spinal Injury Center, Department of Neurological Surgery, University of California, San Francisco, California 94110
| | - Barrett Allen
- Brain and Spinal Injury Center, Department of Neurological Surgery, University of California, San Francisco, California 94110
| | - David Leu
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, California 94304
| | - Susanna Rosi
- Brain and Spinal Injury Center, Department of Neurological Surgery, University of California, San Francisco, California 94110
- Physical Therapy and Rehabilitation Science, University of California, San Francisco, California 94110
| | - Jacob Raber
- Departments of Behavioral Neuroscience, Neurology, and Radiation Medicine, Division of Neuroscience, ONPRC, Oregon Health and Science University, Portland, Oregon 97239
| | - Ting-Ting Huang
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, California 94304
- Geriatric Research, Education and Clinical Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, California 94304
| | - John R. Fike
- Brain and Spinal Injury Center, Department of Neurological Surgery, University of California, San Francisco, California 94110
- Department of Radiation Oncology, University of California, San Francisco, California 94110
| |
Collapse
|
39
|
Rana P, Gupta M, Khan AR, Hemanth Kumar BS, Roy R, Khushu S. NMR based metabolomics reveals acute hippocampal metabolic fluctuations during cranial irradiation in murine model. Neurochem Int 2014; 74:1-7. [PMID: 24787771 DOI: 10.1016/j.neuint.2014.04.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 04/15/2014] [Accepted: 04/21/2014] [Indexed: 01/28/2023]
Abstract
Cranial irradiation is widely used as a treatment modality or prophylactic treatment in cancer patients, but it is frequently related to neurocognitive impairment in cancer survivors. Though most of radiation-induced changes occur during early and late delayed phase of radiation sickness, recent reports have supported the evidence of impaired neurogenesis within 24-48 h of radiation exposure that may implicate changes in acute phase as well. Inspection of these acute changes could be considered important as they may have long lasting effect on cognitive development and functions. In the present study, (1)H NMR spectroscopy based metabolomic approach was used to obtain comprehensive information of hippocampus metabolic physiology during acute phase of radiation sickness in a mouse model for single dose 8 Gy cranial irradiation. The analysis demonstrated reduced metabolic activity in irradiated animals compared to controls, typically evident in citric acid cycle intermediates, glutamine/glutamate and ketone bodies metabolism thus providing strong indication that the hippocampus is metabolically responsive to radiation exposure. The data suggested reduced glucose utilization, altered intermediary and neurotransmitter metabolism in hippocampus tissue extract. To the best of our knowledge this is the first metabolomic study to document cranial irradiation induced acute metabolic changes using in vitro(1)H NMR spectroscopy.
Collapse
Affiliation(s)
- Poonam Rana
- NMR Research Centre, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Mamta Gupta
- NMR Research Centre, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Ahmad Raza Khan
- NMR Research Centre, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - B S Hemanth Kumar
- NMR Research Centre, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Raja Roy
- Centre for Biomedical Magnetic Resonance (CBMR), SGPGIMS Campus, Lucknow, Uttar Pradesh, India
| | - Subash Khushu
- NMR Research Centre, Institute of Nuclear Medicine and Allied Sciences, Delhi, India.
| |
Collapse
|
40
|
Tseng BP, Giedzinski E, Izadi A, Suarez T, Lan ML, Tran KK, Acharya MM, Nelson GA, Raber J, Parihar VK, Limoli CL. Functional consequences of radiation-induced oxidative stress in cultured neural stem cells and the brain exposed to charged particle irradiation. Antioxid Redox Signal 2014; 20:1410-22. [PMID: 23802883 PMCID: PMC3936501 DOI: 10.1089/ars.2012.5134] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
AIMS Redox homeostasis is critical in regulating the fate and function of multipotent cells in the central nervous system (CNS). Here, we investigated whether low dose charged particle irradiation could elicit oxidative stress in neural stem and precursor cells and whether radiation-induced changes in redox metabolism would coincide with cognitive impairment. RESULTS Low doses (<1 Gy) of charged particles caused an acute and persistent oxidative stress. Early after (<1 week) irradiation, increased levels of reactive oxygen and nitrogen species were generally dose responsive, but were less dependent on dose weeks to months thereafter. Exposure to ion fluences resulting in less than one ion traversal per cell was sufficient to elicit radiation-induced oxidative stress. Whole body irradiation triggered a compensatory response in the rodent brain that led to a significant increase in antioxidant capacity 2 weeks following exposure, before returning to background levels at week 4. Low dose irradiation was also found to significantly impair novel object recognition in mice 2 and 12 weeks following irradiation. INNOVATION Data provide evidence that acute exposure of neural stem cells and the CNS to very low doses and fluences of charged particles can elicit a persisting oxidative stress lasting weeks to months that is associated with impaired cognition. CONCLUSIONS Exposure to low doses of charged particles causes a persistent oxidative stress and cognitive impairment over protracted times. Data suggest that astronauts subjected to space radiation may develop a heightened risk for mission critical performance decrements in space, along with a risk of developing long-term neurocognitive sequelae.
Collapse
Affiliation(s)
- Bertrand P Tseng
- 1 Department of Internal Medicine, Duke University Medical Center , Durham, North Carolina
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Allen AR, Eilertson K, Sharma S, Schneider D, Baure J, Allen B, Rosi S, Raber J, Fike JR. Effects of radiation combined injury on hippocampal function are modulated in mice deficient in chemokine receptor 2 (CCR2). Radiat Res 2013; 180:78-88. [PMID: 23772926 DOI: 10.1667/rr3344.1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Chemokines and their receptors play a crucial role in normal brain function as well as in pathological conditions such as injury and disease-associated neuroinflammation. Chemokine receptor-2 (CCR2), which mediates the recruitment of infiltrating and resident microglia to sites of central nervous system (CNS) inflammation, is upregulated by ionizing irradiation and traumatic brain injury. Our objective was to determine if a deficiency in CCR2 and subsequent effects on brain microglia affect neurogenesis and cognitive function after radiation combined injury (RCI). CCR2 knock-out ⁻/⁻ and wild-type (WT) mice received 4 Gy of whole body ¹³⁷Cs irradiation. Immediately after irradiation, unilateral traumatic brain injury was induced using a controlled cortical impact system. Forty-four days postirradiation, animals were tested for hippocampus-dependent cognitive performance in the Morris water-maze. After cognitive testing, animals were euthanized and their brains snap frozen for immunohistochemical assessment of neuroinflammation (activated microglia) and neurogenesis in the hippocampal dentate gyrus. All animals were able to locate the visible and hidden platform locations in the water maze; however, treatment effects were seen when spatial memory retention was assessed in the probe trials (no platform). In WT animals that received combined injury, a significant impairment in spatial memory retention was observed in the probe trial after the first day of hidden platform training (first probe trial). This impairment was associated with increased neurogenesis in the ipsilateral hemisphere of the dentate gyrus. In contrast, CCR2⁻/⁻ mice, independent of insult showed significant memory retention in the first probe trial and there were no differences in the numbers of newly born neurons in the animals receiving irradiation, trauma or combined injury. Although the mechanisms involved are not clear, our data suggests that CCR2 deficiency can exert a protective effect preventing the impairment of cognitive function after combined injury.
Collapse
Affiliation(s)
- Antiño R Allen
- Brain and Spinal Injury Center, Department of Neurological Surgery, University of California, San Francisco, California 94110, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Tseng BP, Lan ML, Tran KK, Acharya MM, Giedzinski E, Limoli CL. Characterizing low dose and dose rate effects in rodent and human neural stem cells exposed to proton and gamma irradiation. Redox Biol 2013; 1:153-62. [PMID: 24024148 PMCID: PMC3757683 DOI: 10.1016/j.redox.2013.01.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 01/07/2013] [Accepted: 01/10/2013] [Indexed: 01/25/2023] Open
Abstract
Past work has shown that exposure to gamma rays and protons elicit a persistent oxidative stress in rodent and human neural stem cells (hNSCs). We have now adapted these studies to more realistic exposure scenarios in space, using lower doses and dose rates of these radiation modalities, to further elucidate the role of radiation-induced oxidative stress in these cells. Rodent neural stem and precursor cells grown as neurospheres and human neural stem cells grown as monolayers were subjected to acute and multi-dosing paradigms at differing dose rates and analyzed for changes in reactive oxygen species (ROS), reactive nitrogen species (RNS), nitric oxide and superoxide for 2 days after irradiation. While acute exposures led to significant changes in both cell types, hNSCs in particular, exhibited marked and significant elevations in radiation-induced oxidative stress. Elevated oxidative stress was more significant in hNSCs as opposed to their rodent counterparts, and hNSCs were significantly more sensitive to low dose exposures in terms of survival. Combinations of protons and γ-rays delivered as lower priming or higher challenge doses elicited radioadaptive changes that were associated with improved survival, but in general, only under conditions where the levels of reactive species were suppressed compared to cells irradiated acutely. Protective radioadaptive effects on survival were eliminated in the presence of the antioxidant N-acetylcysteine, suggesting further that radiation-induced oxidative stress could activate pro-survival signaling pathways that were sensitive to redox state. Data corroborates much of our past work and shows that low dose and dose rate exposures elicit significant changes in oxidative stress that have functional consequences on survival.
Collapse
Key Words
- CM-H2DCFDA (or CM), 5-(and 6-) Chloromethyl-2,7-dichlorodihydrofluorescein diacetate
- DAF, 4-Amino-5methylamino-2′,7′-difluorescein diacetate
- DNA damage
- Dose rate
- GCR, Galactic cosmic rays
- HDR, High dose rate
- LDR, Low dose rate
- LET, Linear energy transfer
- MS, Mitosox
- NAC, N-acetylcysteine
- Neural stem cells
- Oxidative stress
- Protons
- RNS, Reactive nitrogen species
- ROS, Reactive oxygen species
- Radiation
Collapse
Affiliation(s)
- Bertrand P. Tseng
- Department of Internal Medicine, Duke University Medical Center, Durham, NC 27710, United States
| | - Mary L. Lan
- Department of Radiation Oncology, University of California, Irvine, CA 92697-2695, United States
| | - Katherine K. Tran
- Department of Radiation Oncology, University of California, Irvine, CA 92697-2695, United States
| | - Munjal M. Acharya
- Department of Radiation Oncology, University of California, Irvine, CA 92697-2695, United States
| | - Erich Giedzinski
- Department of Radiation Oncology, University of California, Irvine, CA 92697-2695, United States
| | - Charles L. Limoli
- Department of Radiation Oncology, University of California, Irvine, CA 92697-2695, United States
- Correspondence to: Department of Radiation Oncology University of California Irvine, Medical Sciences I, Room B-146B Irvine CA 92697-2695, USA. Tel.: +1 949 824 3053; fax: +1 949 824 3566.
| |
Collapse
|
43
|
Extracellular superoxide dismutase is important for hippocampal neurogenesis and preservation of cognitive functions after irradiation. Proc Natl Acad Sci U S A 2012; 109:21522-7. [PMID: 23236175 DOI: 10.1073/pnas.1216913110] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Cranial irradiation is widely used in cancer therapy, but it often causes cognitive defects in cancer survivors. Oxidative stress is considered a major cause of tissue injury from irradiation. However, in an earlier study mice deficient in the antioxidant enzyme extracellular superoxide dismutase (EC-SOD KO) showed reduced sensitivity to radiation-induced defects in hippocampal functions. To further dissect the role of EC-SOD in neurogenesis and in response to irradiation, we generated a bigenic EC-SOD mouse model (OE mice) that expressed high levels of EC-SOD in mature neurons in an otherwise EC-SOD-deficient environment. EC-SOD deficiency was associated with reduced progenitor cell proliferation in the subgranular zone of dentate gyrus in KO and OE mice. However, high levels of EC-SOD in the granule cell layer supported normal maturation of newborn neurons in OE mice. Following irradiation, wild-type mice showed reduced hippocampal neurogenesis, reduced dendritic spine densities, and defects in cognitive functions. OE and KO mice, on the other hand, were largely unaffected, and the mice performed normally in neurocognitive tests. Although the resulting hippocampal-related functions were similar in OE and KO mice following cranial irradiation, molecular analyses suggested that they may be governed by different mechanisms: whereas neurotrophic factors may influence radiation responses in OE mice, dendritic maintenance may be important in the KO environment. Taken together, our data suggest that EC-SOD plays an important role in all stages of hippocampal neurogenesis and its associated cognitive functions, and that high-level EC-SOD may provide protection against irradiation-related defects in hippocampal functions.
Collapse
|
44
|
Corniola R, Zou Y, Leu D, Fike JR, Huang TT. Paradoxical relationship between Mn superoxide dismutase deficiency and radiation-induced cognitive defects. PLoS One 2012; 7:e49367. [PMID: 23145165 PMCID: PMC3493523 DOI: 10.1371/journal.pone.0049367] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Accepted: 10/10/2012] [Indexed: 02/04/2023] Open
Abstract
Radiation therapy of the CNS, even at low doses, can lead to deficits in neurocognitive functions. Reduction in hippocampal neurogenesis is usually, but not always, associated with cognitive deficits resulting from radiation therapy. Generation of reactive oxygen species is considered the main cause of radiation-induced tissue injuries, and elevated levels of oxidative stress persist long after the initial cranial irradiation. Consequently, mutant mice with reduced levels of the mitochondrial antioxidant enzyme, Mn superoxide dismutase (MnSOD or Sod2), are expected to be more sensitive to radiation-induced changes in hippocampal neurogenesis and the related functions. In this study, we showed that MnSOD deficiency led to reduced generation of immature neurons in Sod2−/+ mice even though progenitor cell proliferation was not affected. Compared to irradiated Sod2+/+ mice, which showed cognitive defects and reduced differentiation of newborn cells towards the neuronal lineage, irradiated Sod2−/+ mice showed normal hippocampal-dependent cognitive functions and normal differentiation pattern for newborn neurons and astroglia. However, we also observed a disproportional decrease in newborn neurons in irradiated Sod2−/+ following behavioral studies, suggesting that MnSOD deficiency may render newborn neurons more sensitive to stress from behavioral trainings following cranial irradiation. A positive correlation between normal cognitive functions and normal dendritic spine densities in dentate granule cells was observed. The data suggest that maintenance of synaptic connections, via maintenance of dendritic spines, may be important for normal cognitive functions following cranial irradiation.
Collapse
Affiliation(s)
- Rikki Corniola
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, United States of America
| | - Yani Zou
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, United States of America
| | - David Leu
- Palo Alto Institute for Research and Education, Palo Alto, California, United States of America
- Geriatric Research, Education, and Clinical Center (GRECC), VA Palo Alto Health Care System, Palo Alto, California, United States of America
| | - John R. Fike
- Departments of Neurosurgery and Radiation Oncology, University of California San Francisco, San Francisco, California, United States of America
| | - Ting-Ting Huang
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, United States of America
- Geriatric Research, Education, and Clinical Center (GRECC), VA Palo Alto Health Care System, Palo Alto, California, United States of America
- * E-mail:
| |
Collapse
|
45
|
Abstract
Changes in the intracellular and extracellular redox balance have been correlated with cell fate decisions in terms of proliferation versus differentiation, entering versus existing cell cycle and survival versus cell death. Adult hippocampal neurogenesis has been correlated with neuronal plasticity of learning and memory; however, the process is exquisitely sensitive to changes in redox balance. Cranial irradiation is an effective modality in treating brain tumours but often leads to deficits in hippocampus-related learning and memory, which is most likely due to sustained elevation of oxygen free radical production and suppression of hippocampal neurogenesis. The subcellular redox environment affecting hippocampal neurogenesis is largely unknown. Using mutant mice deficient in each one of the three superoxide dismutase (SOD, EC 1.15.1.1) isoforms, we have begun to determine the consequences of SOD deficiency in hippocampal neurogenesis and the related functions of learning and memory under normal condition and following cranial irradiation.
Collapse
Affiliation(s)
- Ting-Ting Huang
- Geriatric Research, Education, and Care Center-GRECC, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA.
| |
Collapse
|
46
|
Huang TT, Zou Y, Corniola R. Oxidative stress and adult neurogenesis--effects of radiation and superoxide dismutase deficiency. Semin Cell Dev Biol 2012; 23:738-44. [PMID: 22521481 DOI: 10.1016/j.semcdb.2012.04.003] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Revised: 03/30/2012] [Accepted: 04/04/2012] [Indexed: 01/02/2023]
Abstract
Hippocampus plays an important role in learning and memory and in spatial navigation. Production of new neurons that are functionally integrated into the hippocampal neuronal network is important for the maintenance of functional plasticity. In adults, production of new neurons in the hippocampus takes place in the subgranular zone (SGZ) of dentate gyrus. Neural progenitor/stem cells go through processes of proliferation, differentiation, migration, and maturation. This process is exquisitely sensitive to oxidative stress, and perturbation in the redox balance in the neurogenic microenvironment can lead to reduced neurogenesis. Cranial irradiation is an effective treatment for primary and secondary brain tumors. However, even low doses of irradiation can lead to persistent elevation of oxidative stress and sustained suppression of hippocampal neurogenesis. Superoxide dismutases (SODs) are major antioxidant enzymes for the removal of superoxide radicals in different subcellular compartments. To identify the subcellular location where reactive oxygen species (ROS) are continuously generated after cranial irradiation, different SOD deficient mice have been used to determine the effects of irradiation on hippocampal neurogenesis. The study results suggest that, regardless of the subcellular location, SOD deficiency leads to a significant reduction in the production of new neurons in the SGZ of hippocampal dentate gyrus. In exchange, the generation of new glial cells was significantly increased. The SOD deficient condition, however, altered the tissue response to irradiation, and SOD deficient mice were able to maintain a similar level of neurogenesis after irradiation while wild type mice showed a significant reduction in the production of new neurons.
Collapse
Affiliation(s)
- Ting-Ting Huang
- Geriatric Research, Education, and Care Center (GRECC), VA Palo Alto Health Care System, Palo Alto, CA 94304, USA.
| | | | | |
Collapse
|
47
|
Stessin AM, Gursel DB, Schwartz A, Parashar B, Kulidzhanov FG, Sabbas AM, Boockvar J, Nori D, Wernicke AG. FTY720, sphingosine 1-phosphate receptor modulator, selectively radioprotects hippocampal neural stem cells. Neurosci Lett 2012; 516:253-8. [PMID: 22507238 DOI: 10.1016/j.neulet.2012.04.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Revised: 03/30/2012] [Accepted: 04/01/2012] [Indexed: 12/27/2022]
Abstract
Cranial irradiation is an effective treatment modality for both primary and metastatic brain tumors, yet it induces cognitive decline in a substantial number of patients. At present, there are no established methods for neuroprotection. Recent investigations have revealed a link between radiation-induced cognitive dysfunction and the loss of neural precursor cells in the hippocampus. Hence, identifying pharmacological agents, capable of protecting this cell population, is of interest. FTY720 (fingolimod), an FDA-approved oral drug for the treatment of multiple sclerosis, has been shown to promote the survival and differentiation of neural progenitors, as well as remyelination and repair after brain injury. In this study, we show that FTY720, used at nanomolar concentrations, is capable of increasing the viability and neurogenicity of irradiated neural stem cells from the hippocampus. In contrast, it does not provide radioprotection in a human breast cancer cell line and two glioma cell lines. These results suggest a potential therapeutic role for FTY720 as a neuroprotector during cranial irradiation. Further preclinical studies are warranted to evaluate this possibility.
Collapse
Affiliation(s)
- Alexander M Stessin
- Department of Radiation Oncology, Weill Medical College of Cornell University, New York, NY 10065, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Rosi S, Ferguson R, Fishman K, Allen A, Raber J, Fike JR. The polyamine inhibitor alpha-difluoromethylornithine modulates hippocampus-dependent function after single and combined injuries. PLoS One 2012; 7:e31094. [PMID: 22299052 PMCID: PMC3267765 DOI: 10.1371/journal.pone.0031094] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2011] [Accepted: 01/02/2012] [Indexed: 12/31/2022] Open
Abstract
Exposure to uncontrolled irradiation in a radiologic terrorism scenario, a natural disaster or a nuclear battlefield, will likely be concomitantly superimposed on other types of injury, such as trauma. In the central nervous system, radiation combined injury (RCI) involving irradiation and traumatic brain injury may have a multifaceted character. This may entail cellular and molecular changes that are associated with cognitive performance, including changes in neurogenesis and the expression of the plasticity-related immediate early gene Arc. Because traumatic stimuli initiate a characteristic early increase in polyamine metabolism, we hypothesized that treatment with the polyamine inhibitor alpha-difluoromethylornithine (DFMO) would reduce the adverse effects of single or combined injury on hippocampus structure and function. Hippocampal dependent cognitive impairments were quantified with the Morris water maze and showed that DFMO effectively reversed cognitive impairments after all injuries, particularly traumatic brain injury. Similar results were seen with respect to the expression of Arc protein, but not neurogenesis. Given that polyamines have been found to modulate inflammatory responses in the brain we also assessed the numbers of total and newly born activated microglia, and found reduced numbers of newly born cells. While the mechanisms responsible for the improvement in cognition after DFMO treatment are not yet clear, the present study provides new and compelling data regarding the potential use of DFMO as a potential countermeasure against the adverse effects of single or combined injury.
Collapse
Affiliation(s)
- Susanna Rosi
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, United States of America
- Department of Physical Therapy and Rehabilitation Science, University of California San Francisco, San Francisco, California, United States of America
- Brain and Spinal Injury Center, University of California San Francisco, San Francisco, California, United States of America
| | - Ryan Ferguson
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, United States of America
- Department of Physical Therapy and Rehabilitation Science, University of California San Francisco, San Francisco, California, United States of America
- Brain and Spinal Injury Center, University of California San Francisco, San Francisco, California, United States of America
| | - Kelly Fishman
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, United States of America
- Brain and Spinal Injury Center, University of California San Francisco, San Francisco, California, United States of America
| | - Antino Allen
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, United States of America
- Brain and Spinal Injury Center, University of California San Francisco, San Francisco, California, United States of America
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, Oregon, United States of America
- Department of Neurology, Oregon Health and Science University, Portland, Oregon, United States of America
- Division of Neuroscience, Oregon National Primate Research Center (ONPRC), Oregon Health and Science University, Portland, Oregon, United States of America
| | - John R. Fike
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, United States of America
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California, United States of America
- Brain and Spinal Injury Center, University of California San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
49
|
Yoo DY, Shin BN, Kim IH, Kim W, Kim DW, Yoo KY, Choi JH, Lee CH, Yoon YS, Choi SY, Won MH, Hwang IK. Effects of Cu,Zn-superoxide dismutase on cell proliferation and neuroblast differentiation in the mouse dentate gyrus. Neurochem Res 2011; 37:261-7. [PMID: 21927927 DOI: 10.1007/s11064-011-0605-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Revised: 09/08/2011] [Accepted: 09/10/2011] [Indexed: 12/28/2022]
Abstract
Oxidative stress is one of the most important factors in reducing adult hippocampal neurogenesis in the adult brain. In this study, we observed the effects of Cu,Zn-superoxide dismutase (SOD1) on lipid peroxidation, cell proliferation, and neuroblast differentiation in the mouse dentate gyrus using malondialdehyde (MDA), Ki67, and doublecortin (DCX), respectively. We constructed an expression vector, PEP-1, fused PEP-1 with SOD1, and generated PEP-1-SOD1 fusion protein. We administered PEP-1 and 100 or 500 μg PEP-1-SOD1 intraperitoneally once a day for 3 weeks and sacrificed at 30 min after the last administrations. PEP-1 administration did not change the MDA levels compared to those in the vehicle-treated group, while PEP-1-SOD1 treatment significantly reduced MDA levels compared to the vehicle-treated group. In the PEP-1-treated group, the number of Ki67-positive nuclei was similar to that in the vehicle-treated group. In the 100 μg PEP-1-SOD1-treated group, the number of Ki67-positive nuclei was slightly decreased; however, in the 500 μg PEP-1-SOD1-treated group, Ki67-positive nuclei were decreased to 78.5% of the vehicle-treated group. The number of DCX-positive neuroblasts in the PEP-1-treated group was similar to that in the vehicle-treated group. However, the arborization of DCX-positive neuroblasts was significantly decreased in both the 100 and 500 μg PEP-1-SOD1-treated groups compared to that in the vehicle-treated group. The number of DCX-positive neuroblasts with tertiary dendrites was markedly decreased in the 500 μg PEP-1-SOD1-treated group. These results suggest that a SOD1 supplement to healthy mice may not be necessary to modulate cell proliferation and neuroblast differentiation in the dentate gyrus.
Collapse
Affiliation(s)
- Dae Young Yoo
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, 151-742, South Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Fike JR. Physiopathology of radiation-induced neurotoxicity. Rev Neurol (Paris) 2011; 167:746-50. [PMID: 21889778 DOI: 10.1016/j.neurol.2011.07.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Accepted: 07/26/2011] [Indexed: 10/17/2022]
Abstract
Ionizing irradiation for the treatment of malignant brain tumors has associated with it a risk of inducing serious morphologic and functional deficits. While obvious tissue damage generally occurs after relatively high radiation doses, cognitive impairment can be seen after lower exposures. The mechanisms responsible for cognitive injury are not well understood, but may involve neurogenesis, a process that is affected by microenvironmental factors including oxidative stress and inflammation. In addition, damage to neurons, either directly or through environmental influences may have a profound impact on cognition. The relationships between cellular response, environmental factors and behavior are complex and difficult to study. However, understanding such issues should provide critical information relevant to the development of strategies and approaches to ameliorate or treat radiation-induced injuries that are associated with behavioral performance.
Collapse
Affiliation(s)
- J R Fike
- Brain and Spinal Injury Center, San Francisco General Hospital, 1001 Potrero Avenue, San Francisco, CA 94110, USA
| |
Collapse
|