Radiation-induced astrocyte senescence is rescued by Δ133p53
Jazyk angličtina Země Velká Británie, Anglie Médium print
Typ dokumentu časopisecké články, Research Support, N.I.H., Extramural, práce podpořená grantem
PubMed
30615147
PubMed Central
PMC6422440
DOI
10.1093/neuonc/noz001
PII: 5273763
Knihovny.cz E-zdroje
- Klíčová slova
- IL-6, astrocytes, p53 isoform, radiation-induced brain injury, senescence,
- MeSH
- astrocyty metabolismus účinky záření MeSH
- kraniální ozáření škodlivé účinky MeSH
- kultivované buňky MeSH
- lidé MeSH
- nádorový supresorový protein p53 metabolismus MeSH
- nádory mozku radioterapie MeSH
- protein - isoformy metabolismus MeSH
- radiační poranění metabolismus MeSH
- stárnutí buněk účinky záření MeSH
- Check Tag
- lidé MeSH
- Publikační typ
- časopisecké články MeSH
- práce podpořená grantem MeSH
- Research Support, N.I.H., Extramural MeSH
- Názvy látek
- nádorový supresorový protein p53 MeSH
- protein - isoformy MeSH
- TP53 protein, human MeSH Prohlížeč
BACKGROUND: Cellular senescence and the senescence-associated secretory phenotype (SASP) may contribute to the development of radiation therapy-associated side effects in the lung and blood vessels by promoting chronic inflammation. In the brain, inflammation contributes to the development of neurologic disease, including Alzheimer's disease. In this study, we investigated the roles of cellular senescence and Δ133p53, an inhibitory isoform of p53, in radiation-induced brain injury. METHODS: Senescent cell types in irradiated human brain were identified with immunohistochemical labeling of senescence-associated proteins p16INK4A and heterochromatin protein Hp1γ in 13 patient cases, including 7 irradiated samples. To investigate the impact of radiation on astrocytes specifically, primary human astrocytes were irradiated and examined for expression of Δ133p53 and induction of SASP. Lentiviral expression of ∆133p53 was performed to investigate its role in regulating radiation-induced cellular senescence and astrocyte-mediated neuroinflammation. RESULTS: Astrocytes expressing p16INK4A and Hp1γ were identified in all irradiated tissues, were increased in number in irradiated compared with untreated cancer patient tissues, and had higher labeling intensity in irradiated tissues compared with age-matched controls. Human astrocytes irradiated in vitro also experience induction of cellular senescence, have diminished Δ133p53, and adopt a neurotoxic phenotype as demonstrated by increased senescence-associated beta-galactosidase activity, p16INK4A, and interleukin (IL)-6. In human astrocytes, Δ133p53 inhibits radiation-induced senescence, promotes DNA double-strand break repair, and prevents astrocyte-mediated neuroinflammation and neurotoxicity. CONCLUSIONS: Restoring expression of the endogenous p53 isoform, ∆133p53, protects astrocytes from radiation-induced senescence, promotes DNA repair, and inhibits astrocyte-mediated neuroinflammation.
Department of Neurology Georgetown University Medical Center Washington DC USA
Department of Pathology Georgetown University Medical Center Washington DC USA
Department of Pathology Johns Hopkins Hospital Baltimore Maryland USA
Department of Pathology University of Maryland Baltimore Maryland USA
p53 Laboratory Biomedical Sciences Institutes Singapore
Regional Centre for Applied Molecular Oncology Masaryk Memorial Cancer Institute Brno Czech Republic
Zobrazit více v PubMed
Patchell RA, Tibbs PA, Regine WF, et al. . Postoperative radiotherapy in the treatment of single metastases to the brain: a randomized trial. JAMA. 1998;280(17):1485–1489. PubMed
Fisher BJ, Bauman GS, Leighton CE, Stitt L, Cairncross JG, Macdonald DR. Low-grade gliomas in children: tumor volume response to radiation. Neurosurg Focus. 1998;4(4):e5. PubMed
Gondi V, Pugh SL, Tome WA, et al. . Preservation of memory with conformal avoidance of the hippocampal neural stem-cell compartment during whole-brain radiotherapy for brain metastases (RTOG 0933): a phase II multi-institutional trial. J Clin Oncol. 2014;32(34):3810–3816. PubMed PMC
Rowe LS, Krauze AV, Ning H, Camphausen KA, Kaushal A. Optimizing the benefit of CNS radiation therapy in the pediatric population-PART 1: understanding and managing acute and late toxicities. Oncology (Williston Park). 2017;31(3):182–188. PubMed
Baskar R, Lee KA, Yeo R, Yeoh KW. Cancer and radiation therapy: current advances and future directions. Int J Med Sci. 2012;9(3):193–199. PubMed PMC
Greene-Schloesser D, Robbins ME, Peiffer AM, Shaw EG, Wheeler KT, Chan MD. Radiation-induced brain injury: a review. Front Oncol. 2012;2:73. PubMed PMC
Soussain C, Ricard D, Fike JR, Mazeron JJ, Psimaras D, Delattre JY. CNS complications of radiotherapy and chemotherapy. Lancet. 2009;374(9701):1639–1651. PubMed
Fischer C, Petriccione M, Donzelli M, Pottenger E. Improving care in pediatric neuro-oncology patients: an overview of the unique needs of children with brain tumors. J Child Neurol. 2016;31(4):488–505. PubMed PMC
Schuitema I, de Sonneville L, Kaspers G, et al. . Executive dysfunction 25 years after treatment with cranial radiotherapy for pediatric lymphoid malignancies. J Int Neuropsychol Soc. 2015;21(9):657–669. PubMed
Agbahiwe H, Rashid A, Horska A, et al. . A prospective study of cerebral, frontal lobe, and temporal lobe volumes and neuropsychological performance in children with primary brain tumors treated with cranial radiation. Cancer. 2017;123(1):161–168. PubMed PMC
Redmond KJ, Mahone EM, Terezakis S, et al. . Association between radiation dose to neuronal progenitor cell niches and temporal lobes and performance on neuropsychological testing in children: a prospective study. Neuro Oncol. 2013;15(3):360–369. PubMed PMC
Silber JH, Radcliffe J, Peckham V, et al. . Whole-brain irradiation and decline in intelligence: the influence of dose and age on IQ score. J Clin Oncol. 1992;10(9):1390–1396. PubMed
Maddrey AM, Bergeron JA, Lombardo ER, et al. . Neuropsychological performance and quality of life of 10 year survivors of childhood medulloblastoma. J Neurooncol. 2005;72(3):245–253. PubMed
Demaria M, O’Leary MN, Chang J, et al. . Cellular senescence promotes adverse effects of chemotherapy and cancer relapse. Cancer Discov. 2017;7(2):165–176. PubMed PMC
Eriksson D, Stigbrand T. Radiation-induced cell death mechanisms. Tumour Biol. 2010;31(4):363–372. PubMed
Muñoz-Espín D, Serrano M. Cellular senescence: from physiology to pathology. Nat Rev Mol Cell Biol. 2014;15(7):482–496. PubMed
Baker DJ, Petersen RC. Cellular senescence in brain aging and neurodegenerative diseases: evidence and perspectives. J Clin Invest. 2018;128(4):1208–1216. PubMed PMC
Citrin DE, Shankavaram U, Horton JA, et al. . Role of type II pneumocyte senescence in radiation-induced lung fibrosis. J Natl Cancer Inst. 2013;105(19):1474–1484. PubMed PMC
Wang Y, Boerma M, Zhou D. ionizing radiation-induced endothelial cell senescence and cardiovascular diseases. Radiat Res. 2016;186(2):153–161. PubMed PMC
Turnquist C, Horikawa I, Foran E, et al. . p53 isoforms regulate astrocyte-mediated neuroprotection and neurodegeneration. Cell Death Differ. 2016;23(9):1515–1528. PubMed PMC
Wang C, Ward ME, Chen R, et al. . Scalable production of iPSC-derived human neurons to identify Tau-lowering compounds by high-content screening. Stem Cell Reports. 2017;9(4):1221–1233. PubMed PMC
Sohn JJ, Schetter AJ, Yfantis HG, et al. . Macrophages, nitric oxide and microRNAs are associated with DNA damage response pathway and senescence in inflammatory bowel disease. PLoS One. 2012;7(9):e44156. PubMed PMC
Rayess H, Wang MB, Srivatsan ES. Cellular senescence and tumor suppressor gene p16. Int J Cancer. 2012;130(8):1715–1725. PubMed PMC
Campisi J. Aging, cellular senescence, and cancer. Annu Rev Physiol. 2013;75:685–705. PubMed PMC
Price RE, Langford LA, Jackson EF, Stephens LC, Tinkey PT, Ang KK. Radiation-induced morphologic changes in the rhesus monkey (Macaca mulatta) brain. J Med Primatol. 2001;30(2):81–87. PubMed
Suman S, Rodriguez OC, Winters TA, Fornace AJ Jr, Albanese C, Datta K. Therapeutic and space radiation exposure of mouse brain causes impaired DNA repair response and premature senescence by chronic oxidant production. Aging (Albany NY). 2013;5(8):607–622. PubMed PMC
Lumniczky K, Szatmári T, Sáfrány G. Ionizing radiation-induced immune and inflammatory reactions in the brain. Front Immunol. 2017;8:517. PubMed PMC
Kuilman T, Michaloglou C, Mooi WJ, Peeper DS. The essence of senescence. Genes Dev. 2010;24(22):2463–2479. PubMed PMC
Monje ML, Mizumatsu S, Fike JR, Palmer TD. Irradiation induces neural precursor-cell dysfunction. Nat Med. 2002;8(9):955–962. PubMed
Monje ML, Toda H, Palmer TD. Inflammatory blockade restores adult hippocampal neurogenesis. Science. 2003;302(5651):1760–1765. PubMed
Dong X, Luo M, Huang G, et al. . Relationship between irradiation-induced neuro-inflammatory environments and impaired cognitive function in the developing brain of mice. Int J Radiat Biol. 2015;91(3):224–239. PubMed
Haveman J, Geerdink AG, Rodermond HM. TNF, IL-1 and IL-6 in circulating blood after total-body and localized irradiation in rats. Oncol Rep. 1998;5(3):679–683. PubMed
Lee WH, Sonntag WE, Mitschelen M, Yan H, Lee YW. Irradiation induces regionally specific alterations in pro-inflammatory environments in rat brain. Int J Radiat Biol. 2010;86(2):132–144. PubMed PMC
Salminen A, Ojala J, Kaarniranta K, Haapasalo A, Hiltunen M, Soininen H. Astrocytes in the aging brain express characteristics of senescence-associated secretory phenotype. Eur J Neurosci. 2011;34(1):3–11. PubMed
Saatman KE, Contreras PC, Smith DH, et al. . Insulin-like growth factor-1 (IGF-1) improves both neurological motor and cognitive outcome following experimental brain injury. Exp Neurol. 1997;147(2):418–427. PubMed
Madathil SK, Carlson SW, Brelsfoard JM, Ye P, D’Ercole AJ, Saatman KE. Astrocyte-specific overexpression of insulin-like growth factor-1 protects hippocampal neurons and reduces behavioral deficits following traumatic brain injury in mice. PLoS One. 2013;8(6):e67204. PubMed PMC
Crowe EP, Tuzer F, Gregory BD, et al. . Changes in the transcriptome of human astrocytes accompanying oxidative stress-induced senescence. Front Aging Neurosci. 2016;8:208. PubMed PMC
Fujita K, Mondal AM, Horikawa I, et al. . p53 isoforms delta133p53 and p53beta are endogenous regulators of replicative cellular senescence. Nat Cell Biol. 2009;11(9):1135–1142. PubMed PMC
Sarnat HB. Immunocytochemical markers of neuronal maturation in human diagnostic neuropathology. Cell Tissue Res. 2015;359(1):279–294. PubMed
von Muhlinen N, Horikawa I, Alam F, et al. . p53 isoforms regulate premature aging in human cells. Oncogene. 2018;37(18):2379–2393. PubMed PMC
Yamamoto A, Taki T, Yagi H, et al. . Cell cycle-dependent expression of the mouse Rad51 gene in proliferating cells. Mol Gen Genet. 1996;251(1):1–12. PubMed
Guillaud P, Vermont J, Seigneurin D. Automatic classification of cells in cell cycle phases based on Ki-67 antigen quantification by fluorescence microscopy. Cell Prolif. 1991;24(5):481–491. PubMed
Davis PK, Ho A, Dowdy SF. Biological methods for cell-cycle synchronization of mammalian cells. Biotechniques. 2001;30(6):1322–6, 1328, 1330. PubMed
Mizumatsu S, Monje ML, Morhardt DR, Rola R, Palmer TD, Fike JR. Extreme sensitivity of adult neurogenesis to low doses of X-irradiation. Cancer Res. 2003;63(14):4021–4027. PubMed
García-Matas S, Gutierrez-Cuesta J, Coto-Montes A, et al. . Dysfunction of astrocytes in senescence-accelerated mice SAMP8 reduces their neuroprotective capacity. Aging Cell. 2008;7(5):630–640. PubMed
Blum-Degena D, Müller T, Kuhn W, Gerlach M, Przuntek H, Riederer P. Interleukin-1β and interleukin-6 are elevated in the cerebrospinal fluid of Alzheimer’s and de novo Parkinson’s disease patients. Neurosci Letters. 1995;202(1–2):17–20. PubMed
Mrak RE. Neuropathology and the neuroinflammation idea. J Alzheimers Dis. 2009;18(3):473–481. PubMed
Wang B, Tanaka K, Ji B, et al. . Low-dose total-body carbon-ion irradiations induce early transcriptional alteration without late Alzheimer’s disease-like pathogenesis and memory impairment in mice. J Neurosci Res. 2014;92(7):915–926. PubMed
Abayomi OK. Pathogenesis of cognitive decline following therapeutic irradiation for head and neck tumors. Acta Oncol. 2002;41(4):346–351. PubMed
Pekny M, Pekna M. Astrocyte reactivity and reactive astrogliosis: costs and benefits. Physiol Rev. 2014;94(4):1077–1098. PubMed
Herranz N, Gil J. Mechanisms and functions of cellular senescence. J Clin Invest. 2018;128(4):1238–1246. PubMed PMC
Stephanie P, Simon JB. Double-strand break repair: 53BP1 comes into focus. Nat Rev Mol Cell Biol. 2013;15(1):7. PubMed
Rola R, Raber J, Rizk A, et al. . Radiation-induced impairment of hippocampal neurogenesis is associated with cognitive deficits in young mice. Exp Neurol. 2004;188(2):316–330. PubMed
Yang L, Yang J, Li G, et al. . Pathophysiological responses in rat and mouse models of radiation-induced brain injury. Mol Neurobiol. 2017;54(2):1022–1032. PubMed PMC
Marmary Y, Adar R, Gaska S, et al. . Radiation-induced loss of salivary gland function is driven by cellular senescence and prevented by IL6 modulation. Cancer Res. 2016;76(5):1170–1180. PubMed
Horikawa I, Park KY, Isogaya K, et al. . Δ133p53 represses p53-inducible senescence genes and enhances the generation of human induced pluripotent stem cells. Cell Death Differ. 2017;24(6):1017–1028. PubMed PMC