Show simple item record

Adaptive radiation‐induced epigenetic alterations mitigated by antioxidants

dc.contributor.authorBernal, Autumn J.
dc.contributor.authorDolinoy, Dana C.
dc.contributor.authorHuang, Dale
dc.contributor.authorSkaar, David A.
dc.contributor.authorWeinhouse, Caren
dc.contributor.authorJirtle, Randy L.
dc.date.accessioned2020-03-17T18:32:37Z
dc.date.available2020-03-17T18:32:37Z
dc.date.issued2013-02
dc.identifier.citationBernal, Autumn J.; Dolinoy, Dana C.; Huang, Dale; Skaar, David A.; Weinhouse, Caren; Jirtle, Randy L. (2013). "Adaptive radiation‐induced epigenetic alterations mitigated by antioxidants." The FASEB Journal 27(2): 665-671.
dc.identifier.issn0892-6638
dc.identifier.issn1530-6860
dc.identifier.urihttps://hdl.handle.net/2027.42/154445
dc.publisherThe Federation of American Societies for Experimental Biology
dc.publisherWiley Periodicals, Inc.
dc.subject.otheragouti mice
dc.subject.otherhormesis
dc.subject.otherreactive oxygen species
dc.subject.otherDNA methylation
dc.titleAdaptive radiation‐induced epigenetic alterations mitigated by antioxidants
dc.typeArticle
dc.rights.robotsIndexNoFollow
dc.subject.hlbsecondlevelBiology
dc.subject.hlbtoplevelScience
dc.description.peerreviewedPeer Reviewed
dc.description.bitstreamurlhttps://deepblue.lib.umich.edu/bitstream/2027.42/154445/1/fsb2027002025.pdf
dc.identifier.doi10.1096/fj.12-220350
dc.identifier.sourceThe FASEB Journal
dc.identifier.citedreferenceKovalchuk, O., Burke, P., Besplug, J., Slovack, M., Filkowski, J., and Pogribny, I. ( 2004 ) Methylation changes in muscle and liver tissues of male and female mice exposed to acute and chronic low‐dose X‐ray‐irradiation. Mutat. Res. 548, 75 – 84
dc.identifier.citedreferenceJirtle, R. L., and Skinner, M. K. ( 2007 ) Environmental epigenomics and disease susceptibility. Nat. Rev. Genet. 8, 253 – 262
dc.identifier.citedreferenceYen, T. T., Gill, A. M., Frigeri, L. G., Barsh, G. S., and Wolff, G. L. ( 1994 ) Obesity, diabetes, and neoplasia in yellow A(vy)/‐mice: ectopic expression of the agouti gene. FASEB J. 8, 479 – 488
dc.identifier.citedreferenceWilliams, G., Harrold, J. A., and Cutler, D. J. ( 2000 ) The hypothalamus and the regulation of energy homeostasis: lifting the lid on a black box. Proc. Nutr. Soc. 59, 385 – 396
dc.identifier.citedreferenceWaterland, R. A., and Jirtle, R. L. ( 2003 ) Transposable elements: targets for early nutritional effects on epigenetic gene regulation. Mol. Cell. Biol. 23, 5293 – 5300
dc.identifier.citedreferenceDolinoy, D. C., Weidman, J. R., Waterland, R. A., and Jirtle, R. L. ( 2006 ) Maternal genistein alters coat color and protects Avy mouse offspring from obesity by modifying the fetal epigenome. Environ. Health Perspect. 114, 567 – 572
dc.identifier.citedreferenceDolinoy, D. C., Huang, D., and Jirtle, R. L. ( 2007 ) Maternal nutrient supplementation counteracts bisphenol A‐induced DNA hypomethylation in early development. Proc. Natl. Acad. Sci. U. S. A. 104, 13056 – 13061
dc.identifier.citedreferenceKaminen‐Ahola, N., Ahola, A., Maga, M., Mallitt, K. A., Fahey, P., Cox, T. C., Whitelaw, E., and Chong, S. ( 2010 ) Maternal ethanol consumption alters the epigenotype and the phenotype of offspring in a mouse model. PLoS Genet. 6, e1000811
dc.identifier.citedreferenceMorgan, H. D., Jin, X. L., Li, A., Whitelaw, E., and O’Neill, C. ( 2008 ) The culture of zygotes to the blastocyst stage changes the postnatal expression of an epigentically labile allele, agouti viable yellow, in mice. Biol. Reprod. 79, 618 – 623
dc.identifier.citedreferenceFeil, R., and Fraga, M. F. ( 2011 ) Epigenetics and the environment: emerging patterns and implications. Nat. Rev. Genet. 13, 97 – 109
dc.identifier.citedreferenceWambi, C. O., Sanzari, J. K., Sayers, C. M., Nuth, M., Zhou, Z., Davis, J., Finnberg, N., Lewis‐Wambi, J. S., Ware, J. H., El‐Deiry, W. S., and Kennedy, A. R. ( 2009 ) Protective effects of dietary antioxidants on proton total‐body irradiation‐mediated hematopoietic cell and animal survival. Radiat. Res. 172, 175 – 186
dc.identifier.citedreferenceInstitute of Laboratory Animal Resources ( 1996 ) Guidelines for the Care and Use of Laboratory Animals, National Academy Press, Washington, DC
dc.identifier.citedreferenceGrunau, C., Clark, S. J., and Rosenthal, A. ( 2001 ) Bisulfite genomic sequencing: systematic investigation of critical experimental parameters. Nucleic Acids Res. 29, E65 – 5
dc.identifier.citedreferenceIlnytskyy, Y., Koturbash, I., and Kovalchuk, O. ( 2009 ) Radiation‐induced bystander effects in vivo are epigenetically regulated in a tissue‐specific manner. Environ. Mol. Mutagen. 50, 105 – 113
dc.identifier.citedreferenceKoturbash, I., Boyko, A., Rodriguez‐Juarez, R., McDonald, R. J., Tryndyak, V. P., Kovalchuk, I., Pogribny, I. P., and Kovalchuk, O. ( 2007 ) Role of epigenetic effectors in maintenance of the long‐term persistent bystander effect in spleen in vivo. Carcinogenesis 28, 1831 – 1838
dc.identifier.citedreferenceKoturbash, I., Rugo, R. E., Hendricks, C. A., Loree, J., Thibault, B., Kutanzi, K., Pogribny, I., Yanch, J. C., Engelward, B. P., and Kovalchuk, O. ( 2006 ) Irradiation induces DNA damage and modulates epigenetic effectors in distant bystander tissue in vivo. Oncogene 25, 4267 – 4275
dc.identifier.citedreferenceKoturbash, I., Zemp, F., Kolb, B., and Kovalchuk, O. ( 2011 ) Sex‐specific radiation‐induced microRNAome responses in the hippocampus, cerebellum and frontal cortex in a mouse model. Mutat. Res. 722, 114 – 118
dc.identifier.citedreferenceBairati, I., Meyer, F., Gelinas, M., Fortin, A., Nabid, A., Brochet, F., Mercier, J. P., Tetu, B., Harel, F., Masse, B., Vigneault, E., Vass, S., del Vecchio, P., and Roy, J. ( 2005 ) A randomized trial of antioxidant vitamins to prevent second primary cancers in head and neck cancer patients. J. Natl. Cancer Inst. 97, 481 – 488
dc.identifier.citedreferenceCalabrese, E. J., and Baldwin, L. A. ( 2002 ) Defining hormesis. Hum. Exp. Toxicol. 21, 91 – 97
dc.identifier.citedreferenceCalabrese, E. J. ( 2009 ) The road to linearity: why linearity at low doses became the basis for carcinogen risk assessment. Arch. Toxicol. 83, 203 – 225
dc.identifier.citedreferenceCalabrese, E. ( 2011 ) Improving the scientific foundations for estimating health risks from the Fukushima incident. Proc. Natl. Acad. Sci. U. S. A. 108, 19447 – 19448
dc.identifier.citedreferencePearce, M. S., Salotti, J. A., Little, M. P., McHugh, K., Lee, C., Kim, K. P., Howe, N. L., Ronckers, C. M., Rajaraman, P., Sir Craft, A. W., Parker, L., and de Gonzalez, A. B. ( 2012 ) Radiation exposure from CT scans in childhood and subsequent risk of leukaemia and brain tumours: a retrospective cohort study. Lancet 380, 499 – 505
dc.identifier.citedreferenceNandakumar, V., Vaid, M., Tollefsbol, T. O., and Katiyar, S. K. ( 2011 ) Aberrant DNA hypermethylation patterns lead to transcriptional silencing of tumor suppressor genes in UVB‐exposed skin and UVB‐induced skin tumors of mice. Carcinogenesis 32, 597 – 604
dc.identifier.citedreferenceAnway, M. D., Cupp, A. S., Uzumcu, M., and Skinner, M. K. ( 2005 ) Epigenetic transgenerational actions of endocrine disruptors and male fertility. Science 308, 1466 – 1469
dc.identifier.citedreferenceVaiserman, A. M. ( 2011 ) Hormesis and epigenetics: is there a link? Ageing Res. Rev. 10, 413 – 421
dc.identifier.citedreferenceSchauer, D. A., and Linton, O. W. ( 2009 ) NCRP report no. 160, ionizing radiation exposure of the population of the United States, medical exposure‐are we doing less with more, and is there a role for health physicists? Health Phys. 97, 1 – 5
dc.identifier.citedreferenceSaenko, V., Ivanov, V., Tsyb, A., Bogdanova, T., Tronko, M., Demidchik, Y., and Yamashita, S. ( 2011 ) The Chernobyl accident and its consequences. Clin. Oncol. (R. Coll. Radiol.) 23, 234 – 243
dc.identifier.citedreferenceAkiba, S. ( 2012 ) Epidemiological studies of Fukushima residents exposed to ionising radiation from the Fukushima Daiichi nuclear power plant prefecture‐a preliminary review of current plans. J. Radiol. Prot. 32, 1 – 10
dc.identifier.citedreferenceSanders, C. ( 2009 ) Radiation Hormesis and the Linear‐No‐Threshold Assumption, Springer, New York
dc.identifier.citedreferenceTawa, R., Kimura, Y., Komura, J., Miyamura, Y., Kurishita, A., Sasaki, M. S., Sakurai, H., and Ono, T. ( 1998 ) Effects of X‐ray irradiation on genomic DNA methylation levels in mouse tissues. J. Radiat. Res. 39, 271 – 278
dc.identifier.citedreferenceKoturbash, I., Kutanzi, K., Hendrickson, K., Rodriguez‐Juarez, R., Kogosov, D., and Kovalchuk, O. ( 2008 ) Radiation‐induced bystander effects in vivo are sex specific. Mutat. Res. 642, 28 – 36
dc.identifier.citedreferenceRugo, R. E., Mutamba, J. T., Mohan, K. N., Yee, T., Chaillet, J. R., Greenberger, J. S., and Engelward, B. P. ( 2011 ) Methyltransferases mediate cell memory of a genotoxic insult. Oncogene 30, 751 – 756
dc.identifier.citedreferenceFranco, R., Schoneveld, O., Georgakilas, A. G., and Panayiotidis, M. I. ( 2008 ) Oxidative stress, DNA methylation and carcinogenesis. Cancer Lett. 266, 6 – 11
dc.identifier.citedreferenceDuhl, D. M., Vrieling, H., Miller, K. A., Wolff, G. L., and Barsh, G. S. ( 1994 ) Neomorphic agouti mutations in obese yellow mice. Nat. Genet. 8, 59 – 65
dc.identifier.citedreferenceDolinoy, D. C., Weinhouse, C., Jones, T. R., Rozek, L. S., and Jirtle, R. L. ( 2010 ) Variable histone modifications at the A(vy) metastable epiallele. Epigenetics 5, 637 – 644
dc.owningcollnameInterdisciplinary and Peer-Reviewed


Files in this item

Show simple item record

Remediation of Harmful Language

The University of Michigan Library aims to describe library materials in a way that respects the people and communities who create, use, and are represented in our collections. Report harmful or offensive language in catalog records, finding aids, or elsewhere in our collections anonymously through our metadata feedback form. More information at Remediation of Harmful Language.

Accessibility

If you are unable to use this file in its current format, please select the Contact Us link and we can modify it to make it more accessible to you.