Therapeutic and space radiation exposure of mouse brain causes impaired DNA repair response and premature senescence by chronic oxidant production

Despite recent epidemiological evidences linking radiation exposure and a number of human ailments including cancer, mechanistic understanding of how radiation inflicts long-term changes in cerebral cortex, which regulates important neuronal functions, remains obscure. The current study dissects molecular events relevant to pathology in cerebral cortex of 6 to 8 weeks old female C57BL/6J mice two and twelve months after exposure to a γ radiation dose (2 Gy) commonly employed in fractionated radiotherapy. For a comparative study, effects of 1.6 Gy heavy ion56Fe radiation on cerebral cortex were also investigated, which has implications for space exploration. Radiation exposure was associated with increased chronic oxidative stress, oxidative DNA damage, lipid peroxidation, and apoptosis. These results when considered with decreased cortical thickness, activation of cell-cycle arrest pathway, and inhibition of DNA double strand break repair factors led us to conclude to our knowledge for the first time that radiation caused aging-like pathology in cerebral cortical cells and changes after heavy ion radiation were more pronounced than γ radiation.

[1]  C. Betsholtz,et al.  Intermediate Filament Protein Partnership in Astrocytes* , 1999, The Journal of Biological Chemistry.

[2]  Yukiko Shimizu,et al.  Studies of the Mortality of Atomic Bomb Survivors.Report 12, Part I. Cancer: 1950–1990 , 2012, Radiation research.

[3]  B. Shukitt-Hale,et al.  Brain Signaling and Behavioral Responses Induced by Exposure to 56Fe-Particle Radiation , 2002, Radiation research.

[4]  J. Manfredi,et al.  Cellular senescence and organismal ageing in the absence of p21CIP1/WAF1 in ku80−/− mice , 2009, EMBO reports.

[5]  M. Blagosklonny Cell cycle arrest is not yet senescence, which is not just cell cycle arrest: terminology for TOR-driven aging , 2012, Aging.

[6]  Lijun Jia,et al.  Induction of p21-dependent senescence by an NAE inhibitor, MLN4924, as a mechanism of growth suppression. , 2011, Neoplasia.

[7]  D. Brenner,et al.  Computed tomography--an increasing source of radiation exposure. , 2007, The New England journal of medicine.

[8]  M. Blagosklonny Answering the ultimate question “What is the Proximal Cause of Aging?” , 2012, Aging.

[9]  R. Butler,et al.  Neurocognitive effects of treatment for childhood cancer. , 2006, Mental retardation and developmental disabilities research reviews.

[10]  V. Bohr,et al.  Aging processes, DNA damage, and repair 1 , 1997, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[11]  T. Bateman,et al.  A murine model for bone loss from therapeutic and space-relevant sources of radiation. , 2006, Journal of applied physiology.

[12]  E. Titova,et al.  Exposure to 56Fe-Particle Radiation Accelerates Electrophysiological Alterations in the Hippocampus of APP23 Transgenic Mice , 2010, Radiation research.

[13]  G. Badhwar,et al.  Depth dependence of absorbed dose, dose equivalent and linear energy transfer spectra of galactic and trapped particles in polyethylene and comparison with calculations of models. , 1998, Radiation Research.

[14]  E. Greer,et al.  Signaling networks in aging , 2008, Journal of Cell Science.

[15]  V. Gallo,et al.  Reduced EGFR signaling in progenitor cells of the adult subventricular zone attenuates oligodendrogenesis after demyelination. , 2007, Neuron glia biology.

[16]  L. Deangelis,et al.  Radiation‐induced dementia in patients cured of brain metastases , 1989, Neurology.

[17]  J. Baak,et al.  Phosphohistone H3 expression has much stronger prognostic value than classical prognosticators in invasive lymph node-negative breast cancer patients less than 55 years of age , 2007, Modern Pathology.

[18]  C. Harmer,et al.  Hyper-Fractionated Radiotherapy for Soft Tissue Sarcoma: Results of the Second Study of Hyper-Fractionated Radiotherapy , 1999, Sarcoma.

[19]  J. Fike,et al.  Redox changes induced in hippocampal precursor cells by heavy ion irradiation , 2007, Radiation and environmental biophysics.

[20]  J L Shinn,et al.  Effects of target fragmentation on evaluation of LET spectra from space radiations: implications for space radiation protection studies. , 1996, Radiation measurements.

[21]  J. Yates,et al.  Recapitulation of premature aging with iPSCs from Hutchinson-Gilford progeria syndrome , 2011, Nature.

[22]  Bernard M. Rabin,et al.  A Longitudinal Study of Operant Responding in RatsIrradiated when 2 Months Old , 2005, Radiation research.

[23]  K. Datta,et al.  Reactive oxygen species in health and disease. , 2000, The National medical journal of India.

[24]  L. Verhey,et al.  Evaluation of the clinical applicability of proton beams in definitive fractionated radiation therapy. , 1982, International journal of radiation oncology, biology, physics.

[25]  R. Setlow,et al.  The hazards of space travel , 2003, EMBO reports.

[26]  J. Poirier,et al.  The cholesterol-lowering drug Probucol increases apolipoprotein e production in the hippocampus of aged rats: implications for Alzheimer’s disease , 2003, Neuroscience.

[27]  M. Pecaut,et al.  HZE radiation and dopaminergic modification of startle and prepulse inhibition in mice , 2005, Physiology & Behavior.

[28]  D. Harman Role of Free Radicals in Aging and Disease , 1992, Annals of the New York Academy of Sciences.

[29]  A. Fornace,et al.  Exposure to Heavy Ion Radiation Induces Persistent Oxidative Stress in Mouse Intestine , 2012, PloS one.

[30]  M. Mattson,et al.  Endonuclease VIII-like 1 (NEIL1) promotes short-term spatial memory retention and protects from ischemic stroke-induced brain dysfunction and death in mice , 2012, Proceedings of the National Academy of Sciences.

[31]  Daniele Nosi,et al.  The Neuron-Astrocyte-Microglia Triad in Normal Brain Ageing and in a Model of Neuroinflammation in the Rat Hippocampus , 2012, PloS one.

[32]  Andre Obenaus,et al.  Magnetic Resonance Imaging and Spectroscopy of the Rat Hippocampus 1 Month after Exposure to 56Fe-Particle Radiation , 2008, Radiation research.

[33]  B. Ames,et al.  Oxidants, antioxidants, and the degenerative diseases of aging. , 1993, Proceedings of the National Academy of Sciences of the United States of America.

[34]  M. Frontini,et al.  Interaction between the Cockayne syndrome B and p53 proteins: implications for aging , 2012, Aging.

[35]  D A Pierce,et al.  Studies of the mortality of atomic bomb survivors. Report 12, Part I. Cancer: 1950-1990. , 1996, Radiation research.

[36]  H. Vogel,et al.  Deletion of Ku70, Ku80, or Both Causes Early Aging without Substantially Increased Cancer , 2007, Molecular and Cellular Biology.

[37]  C. Moon,et al.  Increased expression of the embryonic intermediate filament, nestin, in the brains of scrapie-infected mice , 2004, Neuroscience Letters.

[38]  L. Loeb,et al.  DNA damage and repair in brain: relationship to aging. , 1992, Mutation research.

[39]  B. Shukitt-Hale,et al.  Exposure to 16O-Particle Radiation Causes Aging-Like Decrements in Rats through Increased Oxidative Stress, Inflammation and Loss of Autophagy , 2011, Radiation research.

[40]  Li-chun Wei,et al.  Enhancement of nestin protein-immunoreactivity induced by ionizing radiation in the forebrain ependymal regions of rats , 2002, Neuroscience Research.

[41]  R. Redberg,et al.  Cancer risks and radiation exposure from computed tomographic scans: how can we be sure that the benefits outweigh the risks? , 2009, Archives of internal medicine.

[42]  J. Fike,et al.  Radiation Response of Neural Precursor Cells: Linking Cellular Sensitivity to Cell Cycle Checkpoints, Apoptosis and Oxidative Stress , 2004, Radiation research.

[43]  G. Germino,et al.  Rapamycin ameliorates PKD resulting from conditional inactivation of Pkd1. , 2010, Journal of the American Society of Nephrology : JASN.

[44]  Rebecca S Lewis,et al.  Projected cancer risks from computed tomographic scans performed in the United States in 2007. , 2009, Archives of internal medicine.

[45]  M. Blagosklonny,et al.  Rapamycin suppresses brain aging in senescence-accelerated OXYS rats , 2013, Aging.

[46]  J. Fike,et al.  High-LET Radiation Induces Inflammation and Persistent Changes in Markers of Hippocampal Neurogenesis , 2005, Radiation research.

[47]  D. Nam,et al.  Trans-Differentiation of Neural Stem Cells: A Therapeutic Mechanism Against the Radiation Induced Brain Damage , 2012, PloS one.

[48]  Anil Wipat,et al.  Feedback between p21 and reactive oxygen production is necessary for cell senescence , 2010, Molecular systems biology.

[49]  N. Sharpless,et al.  p38MAPK controls expression of multiple cell cycle inhibitors and islet proliferation with advancing age. , 2009, Developmental cell.

[50]  J. Fike,et al.  High-LET Radiation-Induced Response of Microvessels in the Hippocampus , 2010, Radiation research.

[51]  P. Atadja,et al.  Increased activity of p53 in senescing fibroblasts. , 1995, Proceedings of the National Academy of Sciences of the United States of America.

[52]  B. Shukitt-Hale,et al.  Spatial Learning and Memory Deficits Induced by Exposure to Iron-56-Particle Radiation , 2000, Radiation research.

[53]  Bernard M. Rabin,et al.  Effects of age and diet on the heavy particle-induced disruption of operant responding produced by a ground-based model for exposure to cosmic rays , 2005, Brain Research.

[54]  Y. Suh Cell signaling in aging and apoptosis , 2002, Mechanisms of Ageing and Development.

[55]  R. Weinberg,et al.  The signals and pathways activating cellular senescence. , 2005, The international journal of biochemistry & cell biology.

[56]  K. Manda,et al.  Memory impairment, oxidative damage and apoptosis induced by space radiation: Ameliorative potential of α-lipoic acid , 2008, Behavioural Brain Research.

[57]  M. Pecaut,et al.  The Effects of Low-Dose, High-LET Radiation Exposure on Three Models of Behavior in C57BL/6 Mice , 2004, Radiation research.

[58]  G. Murali,et al.  Modulatory role of grape seed extract on age-related oxidative DNA damage in central nervous system of rats , 2006, Brain Research Bulletin.

[59]  J. Roth,et al.  Overexpression of the p21 sdi1 gene induces senescence-like state in human cancer cells: implication for senescence-directed molecular therapy for cancer , 1999, Cell Death and Differentiation.

[60]  Marco Durante,et al.  Heavy ion carcinogenesis and human space exploration , 2008, Nature Reviews Cancer.

[61]  B. Liu,et al.  ROS and p53: a versatile partnership. , 2008, Free radical biology & medicine.

[62]  E. Shaw,et al.  Managing the cognitive effects of brain tumor radiation therapy , 2006, Current treatment options in oncology.

[63]  S. Jackson,et al.  Human cell senescence as a DNA damage response , 2005, Mechanisms of Ageing and Development.

[64]  G. Fiskum,et al.  A role for mitochondrial dysfunction in perpetuating radiation-induced genomic instability. , 2006, Cancer research.

[65]  Richard Weindruch,et al.  Gene expression profiling of aging reveals activation of a p53-mediated transcriptional program , 2007, BMC Genomics.

[66]  K. Manda,et al.  Space radiation‐induced inhibition of neurogenesis in the hippocampal dentate gyrus and memory impairment in mice: ameliorative potential of the melatonin metabolite, AFMK , 2008, Journal of pineal research.

[67]  D. Miglioretti,et al.  Radiation dose associated with common computed tomography examinations and the associated lifetime attributable risk of cancer. , 2009, Archives of internal medicine.

[68]  Yuka Ishida,et al.  Postnatal Changes in Mice Exposed In Utero to Fast Neutrons , 2004 .

[69]  T. Herman,et al.  Low-dose γ-radiation-induced oxidative stress response in mouse brain and gut: regulation by NFκB-MnSOD cross-signaling. , 2011, Mutation research.

[70]  Andre Obenaus,et al.  Hippocampal Neurogenesis and Neuroinflammation after Cranial Irradiation with 56Fe Particles , 2008, Radiation research.

[71]  S. Gatley,et al.  Long-term effects of irradiation with iron-56 particles on the nigrostriatal dopamine system , 2009, Radiation and environmental biophysics.

[72]  H. Ahsan,et al.  Reactive oxygen species: role in the development of cancer and various chronic conditions , 2006, Journal of carcinogenesis.

[73]  J A Corsellis,et al.  VARIATION WITH AGE IN THE VOLUMES OF GREY AND WHITE MATTER IN THE CEREBRAL HEMISPHERES OF MAN: MEASUREMENTS WITH AN IMAGE ANALYSER , 1980, Neuropathology and applied neurobiology.

[74]  H. Withers,et al.  Radiation-induced astrocytic and microglial responses in mouse brain. , 1993, Radiotherapy and oncology : journal of the European Society for Therapeutic Radiology and Oncology.

[75]  Radiation injury and neurogenesis , 2003, Current opinion in neurology.

[76]  F A Cucinotta,et al.  The effects of delta rays on the number of particle-track traversals per cell in laboratory and space exposures. , 1998, Radiation research.

[77]  T. Morgan,et al.  Increased transcription of the astrocyte gene GFAP during middle-age is attenuated by food restriction: implications for the role of oxidative stress. , 1997, Free radical biology & medicine.

[78]  R. Ullrich,et al.  Radiation Leukemogenesis in Mice: Loss of PU.1 on Chromosome 2 in CBA and C57BL/6 Mice after Irradiation with 1 GeV/nucleon 56Fe Ions, X Rays or γ Rays. Part I. Experimental Observations , 2009, Radiation research.

[79]  A. Fornace,et al.  Accelerated hematopoietic toxicity by high energy 56Fe radiation , 2012, International journal of radiation biology.

[80]  K. Datta,et al.  Base Damage Immediately Upstream from Double-Strand Break Ends is a More Severe Impediment to Nonhomologous End Joining than Blocked 3′-Termini , 2011, Radiation research.

[81]  G. Chen,et al.  Current considerations in heavy charged-particle radiotherapy: a clinical research trial of the University of California Lawrence Berkeley Laboratory, Northern California Oncology Group, and Radiation Therapy Oncology Group. , 1985, Radiation research. Supplement.

[82]  Jiri Bartek,et al.  p16INK4A is a robust in vivo biomarker of cellular aging in human skin , 2006, Aging cell.

[83]  W. Engels,et al.  A Third Link connecting Aging with Double Strand Break Repair , 2007, Cell cycle.

[84]  B. Marples,et al.  Persistence of Chromosome Aberrations in Mice Acutely Exposed to 56Fe+26 Ions , 2004, Radiation research.

[85]  J. Campisi Senescent Cells, Tumor Suppression, and Organismal Aging: Good Citizens, Bad Neighbors , 2005, Cell.

[86]  R. Kleinerman Cancer risks following diagnostic and therapeutic radiation exposure in children , 2006, Pediatric Radiology.

[87]  C. Bouras,et al.  Analysis of gial acidic fibrillary protein in the human entorhinal cortex during aging and in Alzheimer's disease , 2003 .

[88]  I. Colin,et al.  Oxidative stress: a required condition for thyroid cell proliferation. , 2010, The American journal of pathology.

[89]  Gloria Dal Forno,et al.  Cognitive decline strongly correlates with cortical atrophy in Alzheimer’s dementia , 1998, Neurobiology of Aging.

[90]  F A Cucinotta,et al.  Applications of amorphous track models in radiation biology , 1999, Radiation and environmental biophysics.