研究業績 / Recent Publications

  1. Shimizu N, Hamada Y, Morozumi R, Yamamoto J, Iwai S, Sugiyama KI, Ide H, Tsuda M: Repair of topoisomerase 1-induced DNA damage by tyrosyl-DNA phosphodiesterase 2 (TDP2) is dependent on its magnesium binding. J Biol Chem. in press doi: 10.1016/j.jbc.2023.104988
  2. 杉山圭一:ゲノム不安定性から見た食品の安全性, 食品加工技術. 2023;43:13-20
  3. Murata Y, Natsume M, Iso T, Shigeta Y, Hirose N, Umano T, Horibata K, Sugiyama KI, Masumura K, Hirose A, Matsumoto M: In vivo mutagenicity assessment of styrene in MutaMouse liver and lung. Genes Environ. 2023; 45:1. doi: 10.1186/s41021-023-00270-9
  4. 杉山圭一, 増村健一:ゲノム不安定性評価を見据えた遺伝毒性研究の新潮流. 日本薬理学雑誌 2022;157:265-70. doi:10.1254/fpj.22015
  5. Hayashi TI, Furuhama A, Yokomizo H, Yamamoto H: Quantitative analyses of misclassification rates in the hazard classification of environmental chemicals: Evaluation of procedures for deriving predicted no-effect concentrations in the Chemical Substance Control Law in Japan. Risk Anal. 2023;43:686-699. doi: 10.1111/risa.13952
  6. Liu W, Yasui M, Sassa A, You X, Wan J, Cao Y, Xi J, Zhang X, Honma M, Luan Y: FTO regulates the DNA damage response via effects on cell-cycle progression. Mutat Res Genet Toxicol Environ Mutagen. 2023;887:503608. doi: 10.1016/j.mrgentox.2023.503608
  7. Honma M, Yamada M, Yasui M, Horibata K, Sugiyama KI , Masumura K: Genotoxicity assessment of food-flavoring chemicals used in Japan. Toxicol Rep. 2022; 9:1008-1012. doi: 10.1016/j.toxrep.2022.04.026
  8. Sugiyama KI, Kinoshita M, Grúz P, Kasamatsu T, Honma M: Bisphenol-A reduces DNA methylation after metabolic activation. Genes Environ. 2022; 44:20. doi: 10.1186/s41021-022-00249-y
  9. Horibata K, Takasawa H, Hojo M, Taquahashi Y, Shigano M, Yokota S, Kobayashi N, Sugiyama KI, Honma M, Hamada S: In vivo genotoxicity assessment of a multiwalled carbon nanotube in a mouse ex vivo culture. Genes Environ. 2022; 44:24. doi: 10.1186/s41021-022-00253-2
  10. Iso T, Natsume M, Murata Y, Shigeta Y, Hirose N, Umano T, Horibata K, Masumura K, Sugiyama K, Matsumoto M, Hirose A: Absence of in vivo mutagenicity of 4,4'-oxybis(benzenesulfonohydrazide) in liver and glandular stomach of Muta™ Mouse. Fundam. Toxicol. Sci. 2022;9:31-36. doi: 10.2131/fts.9.31
  11. Grúz P, Yasui M, Ukai A, Horibata K, Honma M, Sugiyama K: Potent mutagenicity of an azide, 3-azido-1,2-propanediol, in human TK6 cells. Mutat. Res. Genet. Toxicol. Environ. Mutagen. 2022;876-877:503475. doi: 10.1016/j.mrgentox.2022.503475
  12. Petkov PI, Ivanova H, Honma M, Yamada T, Morita T, Furuhama A, Kotov S, Kaloyanova E, Dimitrova G, Mekenyan O: Differences between in vitro and in vivo genotoxicity due to metabolism: The role of kinetics. Computat. Toxicol. 2022;22:100222. doi: 10.1016/j.comtox.2022.100222
  13. Sugiyama K, Kinoshita M, Furusawa H, Sato K, Honma M: Epigenetic effect of the mycotoxin fumonisin B1 on DNA methylation. Mutagenesis. 2021;36:295-301. doi: 10.1093/mutage/geab019
  14. Sassa A, Fukuda T, Ukai A, Nakamura M, Sato R, Fujiwara S, Hirota K, Takeda S, Sugiyama K, Honma M, Yasui M: Follow-up genotoxicity assessment of Ames-positive/equivocal chemicals using the improved thymidine kinase gene mutation assay in DNA repair-deficient human TK6 cells. Mutagenesis. 2021;36:331-338. doi: 10.1093/mutage/geab025
  15. Suzuki T, Sassa A, Grúz P, Gupta RC, Johnson F, Adachi N, Nohmi T: Error-prone bypass patch by a low-fidelity variant of DNA polymerase zeta in human cells. DNA Repair. 2021;100:103052. doi: 10.1016/j.dnarep.2021.103052
  16. Honma M, Yamada M, Yasui M, Horibata K, Sugiyama K, Masumura K: In vivo and in vitro mutagenicity of perillaldehyde and cinnamaldehyde. Genes Environ. 2021;43:30. doi: 10.1186/s41021-021-00204-3
  17. Kasamatsu T, Kitazawa A, Tajima S, Kaneko M, Sugiyama K, Yamada M, Yasui M, Masumura K, Horibata K, Honma M: Development of a new quantitative structure-activity relationship model for predicting Ames mutagenicity of food flavor chemicals using StarDrop™ auto-Modeller™. Genes Environ. 2021;43:16. doi: 10.1186/s41021-021-00182-6
  18. Masumura K, Ando T, Ukai A, Fujiwara S, Yokose S, You X, Suzuki T, Hayashi H, Nohmi T, Takagi H, Honma M: New homozygous gpt delta transgenic rat strain improves an efficiency of the in vivo mutagenicity assay. Genes Environ. 2021;43:25. doi: 10.1186/s41021-021-00195-1
  19. Aoki Y, Ohno M, Matsumoto M, Matsumoto M, Masumura K, Nohmi T, Tsuzuki T: Characteristic mutations induced in the small intestine of Msh2-knockout gpt delta mice. Genes Environ. 2021;43:27. doi: 10.1186/s41021-021-00196-0
  20. Masumura K, Ando T. Toyoda-Hokaiwado N, Ukai A, Nohmi T, Honma M: Comparison of the frequencies of ENU-induced point mutations in male germ cells and inherited germline mutations in their offspring. Genes Environ. 2021;43:43. doi: 10.1186/s41021-021-00216-z
  21. Gajewicz-Skretna A, Furuhama A, Yamamoto H, Suzuki N: Generating accurate in silico predictions of acute aquatic toxicity for a range of organic chemicals: Towards similarity-based machine learning methods. Chemosphere. 2021;280:130681. doi: 10.1016/j.chemosphere.2021.130681
  22. Furuhama A, Hayashi TI, Yamamoto H: Strategy for development of quantitative structure–activity–activity relationship models for chronic fish toxicity: prediction of early-life stage toxicity to Oryzias latipes from acute Daphnia magna toxicity. Jpn. J. Environ. Toxicol. 2021;24:33-42. doi: 10.11403/jset.24.33
  23. Ishii Y, Takasu S, Grúz P, Masumura K, Ogawa K, Nohmi T, Umemura T: The role of DNA polymerase ζ in benzo[a]pyrene-induced mutagenesis in the mouse lung. Mutagenesis. 2021;geab007. doi: 10.1093/mutage/geab007.
  24. Hagio S, Tsuji N, Furukawa S, Takeuchi K, Hayashi S, Kuroda Y, Honma M, Masumura K: Effect of sampling time on somatic and germ cell mutations induced by acrylamide in gpt delta mice. Genes Environ. 2021;43:4. doi: 10.1186/s41021-021-00175-5.
  25. Gajewicz-Skretna A, Gromelski M, Wyrzykowska E, Furuhama A, Yamamoto H, Suzuki N: Aquatic toxicity (Pre)screening strategy for structurally diverse chemicals: global or local classification tree models? Ecotoxicol Environ Saf. 2021;208:111738. doi: 10.1016/j.ecoenv.2020.111738.
  26. Yasui M, Fukuda T, Ukai A, Maniwa J, Imamura T, Hashizume T, Yamamoto H, Shibuya K, Narumi K, Fujiishi Y, Okada E, Fujishima S, Yamamoto M, Otani N, Nakamura M, Nishimura R, Ueda M, Mishima M, Matsuzaki K, Takeiri A, Tanaka K, Okada Y, Nakagawa M, Hamada S, Kajikawa A, Honda H, Adachi J, Misaki K, Ogawa K, Honma M: Weight of evidence approach using a TK gene mutation assay with human TK6 cells for follow-up of positive results in Ames tests: a collaborative study by MMS/JEMS. Genes Environ. 2021;43:7. doi: 10.1186/s41021-021-00179-1.
  27. Chikura S, Kimoto T, Itoh S, Sanada H, Muto S, Horibata K: Standard protocol for the PIGRET assay, a high-throughput reticulocyte Pig-a assay with an immunomagnetic separation, used in the interlaboratory trial organized by the Mammalian Mutagenicity Study Group of the Japanese Environmental Mutagen and Genome Society. Genes Environ. 2021;43:10. doi: 10.1186/s41021-021-00181-7.
  28. Honma M, Kitazawa A, Kasamatsu T, Sugiyama K: Screening for Ames mutagenicity of food flavor chemicals by (quantitative) structure-activity relationship. Genes Environ. 2020;42:32. doi: 10.1186/s41021-020-00171-1.
  29. Van Bossuyt M, Raitano G, Honma M, Van Hoeck E, Vanhaecke T, Rogiers V, Mertens B, Benfenati E: New QSAR models to predict chromosome damaging potential based on the in vivo micronucleus test. Toxicol Lett. 2020;329:80. doi: 10.1016/j.toxlet.2020.04.016.
  30. Masumura K, Yatagai F, Ochiai M, Nakagama H, Nohmi T: Effects of the scid mutation on X-ray-induced deletions in the brain and spleen of gpt delta mice. Genes Environ. 2020;42:19. doi: 10.1186/s41021-020-00158-y.
  31. Chen R, You X, Cao Y, Masumura K, Ando T, Hamada S, Horibata K, Wan J, Xi J, Zhang X, Honma M, Luan Y: Benchmark dose analysis of multiple genotoxicity endpoints in gpt delta mice exposed to aristolochic acid I. Mutagenesis. 2020;geaa034. doi: 10.1093/mutage/geaa034.
  32. Takeishi A, Kogashi H, Odagiri M, Sasanuma H, Takeda S, Yasui M, Honma M, Suzuki T, Kamiya H, Sugasawa K, Ura K, Sassa A: Tyrosyl-DNA phosphodiesterases are involved in mutagenic events at a ribonucleotide embedded into DNA in human cells. PLoS One. 2020;15:e0244790. doi: 10.1371/journal.pone.0244790.
  33. Honma M: An assessment of mutagenicity of chemical substances by (quantitative) structure-activity relationship. Genes Environ. 2020;42:23.doi: 10.1186/s41021-020-00163-1.
  34. Honma M: Report of the Joint Meeting of the 6th Asian Congress on Environmental Mutagens and the 48th Annual Meeting of the Japanese Environmental Mutagen Society, Tokyo, November 18-20, 2019. Genes Environ. 2020;42:30. doi: 10.1186/s41021-020-00170-2.
  35. Ibrahim MA, Yasui M, Saha LK, Sasanuma H, Honma M, Takeda S. Enhancing the sensitivity of the thymidine kinase assay by using DNA repair-deficient human TK6 cells. Environ Mol Mutagen. 2020; 61:602-610. doi: 10.1002/em.22371.
  36. Grúz P, Shimizu M, Sugiyama K, Yamada M, Honma M. Effect of episomally encoded DNA polymerases on chemically induced mutagenesis at the hisG46 target in Ames test. Genes Environ. 2020; 42:14. doi: 10.1186/s41021-020-00154-2.
  37. Masumura, K, Yatagai, F, Ochiai, M, Nakagama H, Nohmi T. Effects of the scid mutation on X-ray-induced deletions in the brain and spleen of gpt delta mice. Genes Environ. 2020; 42:19. doi: 10.1186/s41021-020-00158-y.
  38. Hori HW, Shimoyoshi S, Tanaka Y, Momonami A, Masumura K, Yamada M, Fujii W, Kitagawa Y, Hayashi M. Multiple-endpoint genotoxicity assay for colon carcinogen 1,2-dimethylhydrazine. Mutat Res. 2020; 849:503130. doi: 10.1016/j.mrgentox.2019.503130.
  39. Schoeny R, Cross KP, DeMarini DM, Elespuru R, Hakura A, Levy DD, Williams RV, Zeiger E, Escobar PA, Howe JR, Kato M, Lott J, Moore MM, Simon S, Stankowski LF Jr, Sugiyama K, van der Leede BM. Revisiting the bacterial mutagenicity assays: Report by a workgroup of the International Workshops on Genotoxicity Testing (IWGT). Mutat Res. 2020; 849:503137. doi: 10.1016/j.mrgentox.2020.503137.
  40. Aoki Y, Taniguchi Y, Matsumoto M, Matsumoto M, Ohno M, Masumura K, Sasaki S, Tsuzuki T, Yamamoto M, Nohmi T. Oxidative-stress-driven mutagenesis in the small intestine of the gpt delta mouse induced by oral administration of potassium bromate. Mutat Res. 2020; 850-851:503136. doi: 10.1016/j.mrgentox.2020.503136.
  41. Martus HJ, Froetschl R, Gollapudi B, Honma M, Marchetti F, Pfuhler S, Schoeny R, Uno Y, Yauk C, Kirkland DJ. Summary of major conclusions from the 7th International Workshop on Genotoxicity Testing (IWGT), Tokyo, Japan. Mutat Res. 2020; 852:503134. doi: 10.1016/j.mrgentox.2020.503134.
  42. 本間 正充. 食品中に混在する微量な化学物質の安全性評価 : 定量的構造活性相関(QSAR)による変異原性化学物質の同定(特集 食品用容器包装の安全性規制(2)食品衛生法改正). 日本包装学会誌. 2020; 29:27-42.
  43. 増村健一. 衛生試験法・注解2020,1.3 遺伝毒性試験法,(公社)日本薬学会編,金原出版(株),東京,pp.135-138, 161-162, 164-166 (2020) (編集および執筆分担).
  44. 堀端克良. 衛生試験法・注解2020,1.3 遺伝毒性試験法,(公社)日本薬学会編,金原出版(株),東京,pp.166-167 (2020).
  45. Sassa A, Tada H, Takeishi A, Harada K, Suzuki M, Tsuda M, Sasanuma H, Takeda S, Sugasawa K, Yasui M, Honma M, Ura K. Processing of a single ribonucleotide embedded into DNA by human nucleotide excision repair and DNA polymerase η. Scientific Reports. 2019; 9:13910. doi: 10.1038/s41598-019-50421-8.
  46. Shemansky JM, McDaniel LP, Klimas C, Dertinger SD, Dobrovolsky VN, Kimoto T, Horibata K, Polli JE, Heflich RH. Pig-a gene mutation database. Environ Mol Mutagen. 2019; 60:759-762. doi: 10.1002/em.22298.
  47. Petkov PI, Kuseva C, Kotov S, Honma M, Kitazawa A, Kulkarni S, Schultz TW, Mekenyan OG. Procedure for toxicological predictions based on mechanistic weight of evidences: Application to Ames mutagenicity. Comput. Toxicol. 2019; 12:100009. doi: 10.1016/j.comtox.2017.02.004.
  48. Aoki Y, Matsumoto M, Matsumoto M, Masumura K, Nohmi T. Mutant frequency is not increased in mice orally exposed to sodium dichromate. Food Safety. 2019; 7:2-10. doi: 10.14252/foodsafetyfscj.2018014.
  49. Sassa A, Yasui M, Honma M. Current perspectives on mechanisms of ribonucleotide incorporation and processing in mammalian DNA. Genes Environ. 2019 Jan 25; 41:3. doi: 10.1186/s41021-019-0118-7.
  50. Chikura S, Kimoto T, Itoh S, Sanada H, Muto S, Horibata K. Standard protocol for the total red blood cell Pig-a assay used in the interlaboratory trial organized by the Mammalian Mutagenicity Study Group of the Japanese Environmental Mutagen Society. Genes Environ. 2019; 41:5. doi: 10.1186/s41021-019-0121-z.
  51. Grúz P, Sugiyama K, Honma M, Nohmi T. Purification and Interactions of the MucA' and MucB Proteins Constituting the DNA Polymerase RI. Genes Environ. 2019; 41:10. doi: 10.1186/s41021-019-0125-8.
  52. Sassa A, Fukuda T, Ukai A, Nakamura M, Takabe M, Takamura-Enya T, Honma M, Yasui M. Comparative study of cytotoxic effects induced by environmental genotoxins using XPC- and CSB-deficient human lymphoblastoid TK6 cells. Genes Environ. 2019; 41:15. doi: 10.1186/s41021-019-0130-y.
  53. Horibata K, Sekimoto M, Sugiyama K. Comprehensive framework between environment and genomic stability: the open symposium of the Japanese Environmental Mutagen Society (JEMS) in 2019. Genes Environ. 2019; 41:17. doi: 10.1186/s41021-019-0132-9.
  54. Sugiyama K, Furusawa H, Grúz P, Kinoshita M, Honma M. Inhibitory effect of ochratoxin A on DNMT-mediated flocculation of yeast. Mutagenesis. 2019; 34:173-180 doi: 10.1093/mutage/gez002.
  55. Gi M, Fujioka M, Totsuka Y, Matsumoto M, Masumura K, Kakehashi A, Yamaguchi T, Fukushima S, Wanibuchi H. Quantitative analysis of mutagenicity and carcinogenicity of 2-amino-3-methylimidazo[4,5-f]quinoline in F344 gpt delta transgenic rats.Mutagenesis. 2019; 34:279-287 doi: 10.1093/mutage/gez015.
  56. Hori H, Shimoyoshi S, Tanaka Y, Momonami A, Masumura K, Yamada M, Fujii W, Kitagawa Y. Integration of micronucleus tests with a gene mutation assay in F344 gpt delta transgenic rats using benzo[a]pyrene. Mutat Res. 2019; 837:1-7. doi: 10.1016/j.mrgentox.2018.09.003.
  57. Dertinger SD, Totsuka Y, Bielas JH, Doherty AT, Kleinjans J, Honma M, Marchetti F, Schuler MJ, Thybaud V, White P, Yauk CL. High information content assays for genetic toxicology testing: A report of the International Workshops on Genotoxicity Testing (IWGT). Mutat Res. 2019; 847:403022. doi: 10.1016/j.mrgentox.2019.02.003.
  58. Lynch AM, Eastmond D, Elhajouji A, Froetschl R, Kirsch-Volders M, Marchetti F, Masumura K, Pacchierotti F, Schuler M, Tweats D. Targets and mechanisms of chemically induced aneuploidy. Part 1 of the report of the 2017 IWGT workgroup on assessing the risk of aneugens for carcinogenesis and hereditary diseases. Mutat Res. 2019; 847:403025. doi: 10.1016/j.mrgentox.2019.02.006.
  59. Tweats D, Eastmond DA, Lynch AM, Elhajouji A, Froetschl R, Kirsch-Volders M, Marchetti F, Masumura K, Pacchierotti F, Schuler M. Role of aneuploidy in the carcinogenic process: Part 3 of the report of the 2017 IWGT workgroup on assessing the risk of aneugens for carcinogenesis and hereditary diseases. Mutat Res. 2019; 847:403032. doi: 10.1016/j.mrgentox.2019.03.005.
  60. Kirkland D, Uno Y, Luijten M, Beevers C, van Benthem J, Burlinson B, Dertinger S, Douglas GR, Hamada S, Horibata K, Lovell DP, Manjanatha M, Martus HJ, Mei N, Morita T, Ohyama W, Williams A. In vivo genotoxicity testing strategies: Report from the 7th International workshop on genotoxicity testing (IWGT). Mutat Res. 2019; 847:403035. doi: 10.1016/j.mrgentox.2019.03.008.
  61. Pacchierotti F, Masumura K, Eastmond DA, Elhajouji A, Froetschl R, Kirsch-Volders M, Lynch A, Schuler M, Tweats D, Marchetti F. Chemically induced aneuploidy in germ cells. Part II of the report of the 2017 IWGT workgroup on assessing the risk of aneugens for carcinogenesis and hereditary diseases. Mutat Res. 2019; 848:403023. doi: 10.1016/j.mrgentox.2019.02.004.
  62. Gollapudi BB, White PA, Honma M. The IWGT in vitro Mammalian Cell Gene Mutation (MCGM) assays working group-Introductory remarks & consensus statements. Mutat Res. 2019; 848:403061. doi: 10.1016/j.mrgentox.2019.05.017.
  63. Levy DD, Zeiger E, Escobar PA, Hakura A, van der Leede BM, Kato M, Moore MM, Sugiyama K. Recommended criteria for the evaluation of bacterial mutagenicity data (Ames test). Mutat Res. 2019; 848:403074. doi: 10.1016/j.mrgentox.2019.07.004.
  64. Levy DD, Hakura A, Elespuru RK, Escobar PA, Kato M, Lott J, Moore MM, Sugiyama K. Demonstrating laboratory proficiency in bacterial mutagenicity assays for regulatory submission. Mutat Res. 2019; 848:403075. doi: 10.1016/j.mrgentox.2019.07.005.
  65. Sugiyama K, Furusawa H, Honma M. Detection of Epigenetic Effects of Citrinin Using a Yeast-Based Bioassay. Mycotoxin Res. 2019; 35:363-368. doi: 10.1007/s12550-019-00361-z.
  66. Hasselgren C, Ahlberg E, Akahori Y, Amberg A, Anger LT, Atienzar F, Auerbach S, Beilke L, Bellion P, Benigni R, Bercu J, Booth ED, Bower D, Brigo A, Cammerer Z, Cronin MTD, Crooks I, Cross KP, Custer L, Dobo K, Doktorova T, Faulkner D, Ford KA, Fortin MC, Frericks M, Gad-McDonald SE, Gellatly N, Gerets H, Gervais V, Glowienke S, Van Gompel J, Harvey JS, Hillegass J, Honma M, Hsieh JH, Hsu CW, Barton-Maclaren TS, Johnson C, Jolly R, Jones D, Kemper R, Kenyon MO, Kruhlak NL, Kulkarni SA, Kümmerer K, Leavitt P, Masten S, Miller S, Moudgal C, Muster W, Paulino A, Lo Piparo E, Powley M, Quigley DP, Reddy MV, Richarz AN, Schilter B, Snyder RD, Stavitskaya L, Stidl R, Szabo DT, Teasdale A, Tice RR, Trejo-Martin A, Vuorinen A, Wall BA, Watts P, White AT, Wichard J, Witt KL, Woolley A, Woolley D, Zwickl C, Myatt GJ. Genetic toxicology in silico protocol. Regul Toxicol Pharmacol. 2019; 107:104403. doi: 10.1016/j.yrtph.2019.104403.
  67. 本間正充. 医薬品中の変異原性不純物の安全性評価と管理―ICH-M7を踏まえた遺伝毒性物質の許容値の設定に関する科学―. PHARM TECH JAPAN. 2019; 35: 1461-1469.
  68. 本間正充. 化学物質の遺伝毒性評価と定量的構造相関((Q)SAR). ポリ衛協会報. 2019; 65; 5-25.
  69. Aoki Y, Nakajima D, Matsumoto M, Yagishita M, Matsumoto M, Yanagisawa R, Goto S, Masumura K, Nohmi T. Change over time of the mutagenicity in the lungs of gpt delta transgenic mice by extract of airborne particles collected from ambient air in the Tokyo metropolitan area. Genes Environ. 2018; 40:25. doi: 10.1186/s41021-018-0113-4.
  70. Grúz P, Shimizu M, Yamada M, Sugiyama K, Honma M. Opposing roles of Y-family DNA polymerases in lipid peroxide mutagenesis at the hisG46 target in the Ames test. Mutat Res Genet Toxicol Environ Mutagen. 2018; 829-830:43-49. doi: 10.1016/j.mrgentox.2018.04.003.
  71. 本間正充. 食品中に含まれる化学物質のリスクアセスメントと遺伝毒性評価 FFI ジャーナル. 2018;223 (1):8-16.
  72. Yasui M, Muto S, Sassa A. Challenges of young scientists at the cutting-edge of genotoxicity research: the open symposium of the Japanese Environmental Mutagen Society (JEMS), 2018. Genes and Environment. 2018; 40:22. doi: 10.1186/s41021-018-0111-6
  73. Mishima M, Hashizume T, Haranosono Y, Nagato Y, Takeshita K, Fukuchi J and Homma M. Meeting report, ICH M7 relevant workshop: use of (Q)SAR systems and expert judgment. Genes and Environment. 2018; 40:19. doi: 10.1186/s41021-018-0107-2
  74. Tennant RE, Guesné SJ, Canipa S, Cayley A, Drewe WC, Honma M, Masumura K, Morita T, Stalford SA, Williams RV. Extrapolation of in vitro structural alerts for mutagenicity to the in vivo endpoint. Mutagenesis. 2019;34:111-121. doi: 10.1093/mutage/gey030.
  75. Amberg A, Anger LT, Bercu J, Bower D, Cross KP, Custer L, Harvey JS, Hasselgren C, Honma M, Johnson C, Jolly R, Kenyon MO, Kruhlak NL, Leavitt P, Quigley DP, Miller S, Snodin D, Stavitskaya L, Teasdale A, Trejo-Martin A, White  AT, Wichard J, Myatt GJ. Extending (Q)SARs to incorporate proprietary knowledge for regulatory purposes: is aromatic N-oxide a structural alert for predicting DNA-reactive mutagenicity? Mutagenesis. 2019;34:67-82. doi: 10.1093/mutage/gey020.
  76. Kohara A, Matsumoto M, Hirose A, Hayashi M, Honma M, Suzuki T. Mutagenic properties of dimethylaniline isomers in mice as evaluated by comet, micronucleus and transgenic mutation assays. Genes Environ. 2018 Aug 22;40:18. doi: 10.1186/s41021-018-0106-3. eCollection 2018.
  77. Morita T, Shigeta Y, Kawamura T, Fujita Y, Honda H, Honma M. In silico prediction of chromosome damage: comparison of three (Q)SAR models. Mutagenesis. 2018 Jul 28. doi: 10.1093/mutage/gey017.
  78. Benfenati E, Golbamaki A, Raitano G, Roncaglioni A, Manganelli S, Lemke F, Norinder U, Lo Piparo E, Honma M, Manganaro A, Gini G. A large comparison of integrated SAR/QSAR models of the Ames test for mutagenicity. SAR QSAR Environ Res. 2018 Aug;29(8):591-611. doi: 10.1080/1062936X.2018.1497702.
  79. Saha LK, Kim S, Kang H, Akter S, Choi K, Sakuma T, Yamamoto T, Sasanuma H, Hirota K, Nakamura J, Honma M, Takeda S, Dertinger S. Differential micronucleus frequency in isogenic human cells deficient in DNA repair pathways is a valuable  indicator for evaluating genotoxic agents and their genotoxic mechanisms. Environ Mol Mutagen. 2018 Jul;59(6):529-538. doi: 10.1002/em.22201.
  80. Myatt GJ, Ahlberg E, Akahori Y, Allen D, Amberg A, Anger LT, Aptula A, Auerbach S, Beilke L, Bellion P, Benigni R, Bercu J, Booth ED, Bower D, Brigo A, Burden N, Cammerer Z, Cronin MTD, Cross KP, Custer L, Dettwiler M, Dobo K, Ford KA, Fortin MC, Gad-McDonald SE, Gellatly N, Gervais V, Glover KP Glowienke S, Van Gompel J, Gutsell S, Hardy B, Harvey JS, Hillegass J, Honma M, Hsieh JH, Hsu CW, Hughes K, Johnson C, Jolly R, Jones D, Kemper R, Kenyon MO, Kim MT, Kruhlak NL, Kulkarni SA, Kümmerer K, Leavitt P, Majer B, Masten S, Miller S, Moser J, Mumtaz M, Muster W, Neilson L, Oprea TI, Patlewicz G, Paulino A, Lo Piparo E, Powley M, Quigley DP, Reddy MV, Richarz AN, Ruiz P, Schilter B, Serafimova R, Simpson W, Stavitskaya L, Stidl R, Suarez-Rodriguez D, Szabo DT, Teasdale A, Trejo-Martin A, Valentin JP, Vuorinen A, Wall BA, Watts P, White AT, Wichard J, Witt KL, Woolley A, Woolley D, Zwickl C, Hasselgren C. In silico toxicology protocols. Regul Toxicol Pharmacol. 2018 Jul;96:1-17. doi: 10.1016/j.yrtph.2018.04.014.
  81. Suzuki T, Matsumoto K, Honma M, Nohmi T. Impact of DNA polymerase ζ mutations on genotoxic thresholds of oxidative mutagens. Mutat Res. 2018, 828:10-14. doi: 10.1016/j.mrgentox.2018.02.001.
  82. You X, Ando T, Xi J, Cao Y, Liu W, Zhang X, Honma M, Masumura K, Luan Y. Gene mutation and micronucleus assays in gpt delta mice treated with 2,2',4,4'-tetrabromodiphenyl ether. Mutagenesis. 2018;33:153-160. doi: 10.1093/mutage/gey002.
  83. Kato M, Sugiyama, K., Fukushima, T., Miura, Y., Awogi, T., Hikosaka, S., Kawakami, K., Nakajima, M., Nakamura, M., Sui, H., Watanabe, K. and Hakura, A: Negative and positive control ranges in the bacterial reverse mutation test: JEMS/BMS collaborative study, Genes and Environ. 40, 1-13 (2018).
  84. Gi M, Fujioka M, Kakehashi A, Okuno T, Masumura K, Nohmi T, Matsumoto M, Omori M, Wanibuchi H, Fukushima S.: In vivo positive mutagenicity of 1,4-dioxane and quantitative analysis of its mutagenicity and carcinogenicity in rats.Arch Toxicol. 2018 Oct;92(10):3207-3221. doi: 10.1007/s00204-018-2282-0.
  85. Masumura K, Toyoda-Hokaiwado N, Niimi N, Grúz P, Wada NA, Takeiri A, Jishage KI, Mishima M, Nohmi T.Limited ability of DNA polymerase kappa to suppress benzo[a]pyrene-induced genotoxicity in vivo.Environ Mol Mutagen. 2017 Dec;58(9):644-653. doi: 10.1002/em.22146.
  86. Nohmi T, Masumura K, Toyoda-Hokaiwado N.Transgenic rat models for mutagenesis and carcinogenesis.Genes Environ. 2017 Feb 1;39:11. doi: 10.1186/s41021-016-0072-6. eCollection 2017.
  87. Sugiyama KI, Furusawa H, Grúz P, Honma M. Functional role of DNA methylation at the FLO1 promoter in budding yeast. FEMS Microbiol Lett. 2017, 1;364(22). doi: 10.1093/femsle/fnx221.
  88. Petkov, PI, Schultz TW, Honma M, Kirilov K, Kotov S, Mekenyan OG. Predicting in vitro genotoxicity by mouse lymphoma L5178Y thymidine kinase mutation assay (MLA): Accounting for simulated metabolic activation of chemicals. Computational Toxicology. 2017, 4, 45-53. Doi:10.1016/j-comtox.2017.10.002
  89. Gadaleta D, Porta N, Vrontaki E, Manganelli S, Manganaro A, Sello G, Honma M, Benfenati E. Integrating computational methods to predict mutagenicity of aromatic azo compounds. J Environ Sci Health C Environ Carcinog Ecotoxicol Rev. 2017, 35(4):239-257. doi: 10.1080/10590501.2017.1391521.
  90. Grúz P, Shimizu M, Sugiyama KI, Honma M. Mutagenicity of ω-3 fatty acid peroxidation products in the Ames test. Mutat Res. 2017 Jul;819:14-19. doi:10.1016/j.mrgentox.2017.05.004.
  91. Sugiyama KI, Furusawa H, Grúz P, Honma M. Detection of epigenetic mutagens including anthracene-derived compounds using yeast FLO1 promoter GFP reporter gene assay. Mutagenesis. 2017 Jul 1;32(4):429-435. doi: 10.1093/mutage/gex009.
  92. Kanemaru Y, Suzuki T, Sassa A, Matsumoto K, Adachi N, Honma M, Numazawa S, Nohmi T. DNA polymerase kappa protects human cells against MMC-induced genotoxicity through error-free translesion DNA synthesis. Genes Environ. 2017 Jan 7;39:6. doi: 10.1186/s41021-016-0067-3. eCollection 2017
  93. Masumura K, Masuda S: Research on environmental mutagenesis from young scientists - the open symposium of the Japanese Environmental Mutagen Society (JEMS) in 2017.Genes and Environment 2017;39:26. doi 10.1186/s41021-017-0086-8
  94. Wada K, Katoh Y, Ohnuma-Koyama A, Takahashi N, Yamada M, Matsumoto K: 2-Nitroanisole-induced oxidative DNA damage in Salmonella typhimurium and in rat urinary bladder cells. Mutat Res; 816-7:18-23 (2017).
  95. Horibata K, Ukai A, Ogata A, Nakae D, Ando H, Kubo Y, Nagasawa A, Yuzawa K, Honma M. Absence of in vivo mutagenicity of multi-walled carbon nanotubes in single intratracheal instillation study using F344 gpt delta rats. Genes Environ. 39, 4 (2017)
  96. 本間正充、森田健 OECD遺伝毒性ガイドラインの改定 国立医薬品食品衛生研究所報告 134: 22-32 (2016)
  97. Grúz P. Lipid Peroxidation, Diet and the Genotoxicology of Aging. In:Malavolta M, Mocchegiani E, editors. Molecular Basis of Nutrition and Aging, First Edition. Elsevier Inc.; 2016. p. 155–76. doi:10.1016/B978- 0-12-801816-3.00001-7.
  98. Yamada M. Preface. Mutat Res. 811, 1-2 (2016)
  99. Kimoto T, Horibata K, Miura D, Chikura S, Okada Y, Ukai A, Itoh S, Nakayama S, Sanada H, Koyama N, Muto S, Uno Y, Yamamoto M, Suzuki Y, Fukuda T, Goto K, Wada K, Kyoya T, Shigano M, Takasawa H, Hamada S, Adachi H, Uematsu Y, Tsutsumi D, Hori H, Kikuzuki R, Ogiwara Y, Yoshida I, Maeda A, Narumi K, Fujiishi Y, Morita T, Yamada M, Honma M. The PIGRET assay, a method for measuring Pig-a gene mutation in reticulocytes, is reliable as a short-term in vivo genotoxicity test: Summary of the MMS/JEMS-collaborative study across 16 laboratories using 24 chemicals. Mutat Res. 811, 3-15 (2016)
  100. Horibata K, Ukai A, Honma M., Evaluation of mutagenicity of acrylamide using RBC Pig-a and PIGRET assays by single peroral dose in rats. Mutat Res. 811, 54-59 (2016)
  101. Johnson GE, Yamamoto M, Suzuki Y, Adachi H, Kyoya T, Takasawa H, Horibata K, Tsutsumi E, Wada K, Kikuzuki R, Yoshida I, Kimoto T, Maeda A, Narumi K. Measuring reproducibility of dose response data for the Pig-a assay using covariate benchmark dose analysis. Mutat Res. 811, 135-139 (2016)
  102. Horibata K, Ukai A, Ishikawa S, Sugano A, Honma M. Monitoring genotoxicity in patients receiving chemotherapy for cancer: application of the PIG-A assay. Mutat Res. 808, 20-26 (2016)
  103. Masumura K, Toyoda-Hokaiwado N, Ukai A, Gondo Y, Honma M, Nohmi T. Dose-dependent de novo germline mutations detected by whole-exome sequencing in progeny of ENU-treated male gpt delta mice. Mutat Res. 810, 30-39 (2016)
  104. Suzuki T, Yasui M, Honma M. Mutator phenotype and DNA double-strand break repair in BLM helicase-deficient human cells. Mol Cell Biol. 36, 2877-2889 (2016)
  105. Suzuki T, Grúz P, Honma M, Adachi N, Nohmi T. The role of DNA polymerase ζ in translesion synthesis across bulky DNA adducts and cross-links in human cells.Mutat Res. 791-792, 35-41 (2016)
  106. Sassa A, Kanemaru Y, Kamoshita N, Honma M, Yasui M. Mutagenic consequences of cytosine alterations site-specifically embedded in the human genome. Genes Environ. 38,17 (2016).
  107. Cao Y, Yang L, Feng N, Shi O, Xi J, You X, Yin C, Yang H, Horibata K, Honma M, Qian B, Weng W, Luan Y. A population study using the human erythrocyte PIG-A assay. Environ Mol Mutagen. 57, 605-614 (2016)
  108. Sugiyama KI, Furusawa H, Shimizu M, Grúz P, Honma M. Epigenetic mutagen as histone modulator can be detected by yeast flocculation. Mutagenesis. 31, 687-693 (2016)
  109. Manganelli S, Benfenati E, Manganaro A, Kulkarni S, Barton-Maclaren TS, Honma M. New quantitative structure-activity relationship models improve predictability of Ames mutagenicity for aromatic azo compounds.Toxicol Sci. 153, 316-26 (2016)
  110. 本間正充 ゲノム上に起きたDNA損傷の運命をターゲットミュータジェネシスにより追跡する.日本がん疫学・分子疫学研究会 News Letter 114, 11-12 (2016)
  111. 本間正充 食品添加物等の遺伝毒性リスク評価法.食品衛生学雑誌 57(1)J12-J15 (2016)
  112. Suzuki T, Grúz P, Honma M, Adachi N, Nohmi T. Sensitivity of human cells expressing low-fidelity or weak-catalytic-activity variants of DNA polymerase ζ to genotoxic stresses.DNA Repair (Amst) 45. 34-43 (2016)
  113. Petko I. Petkov, Terry W. Schultz, E. Maria Donner, Masamitsu Honma, Takeshi Morita, Shuichi Hamada, Akihiro Wakata, Masayuki Mishima, Jiro Maniwa, Milen Todorov, Elena Kaloyanova, Stefan Kotov, Ovanes G. Mekenyan, Integrated Approach to Testing and Assessment for Predicting Rodent Genotoxic Carcinogenesity.Journal of Applied Toxicology 36, 1536-1550 (2016)
  114. E. Benfenati, M. Belli, T. Borges, E. Casimiro, J. Cester, A. Fernandez, G. Gini, M. Honma, M. Kinzl, R. Knauf, A. Manganaro, E. Mombelli, M. I. Petoumenou, M. Paparella, P. Paris & G. Raitano, Results of a round-robin exercise on read-across. SAR and QSAR in Environmental Research 27, 371-384 (2016)
  115. Masumura K, Toyoda-Hokaiwado N, Ukai A, Gondo Y, Honma M, Nohmi T. Estimation of the frequency of inherited germline mutations by whole exome sequencing in ethyl nitrosourea-treated and untreated gpt delta mice. Gene & Environ. 38, 10 (2016)
  116. Morita T, Hamada S, Masumura K, Wakata A, Maniwa J, Takasawa H, Yasunaga K, Hashizume T, Honma M. Evaluation of the sensitivity and specificity of in vivo erythrocytemicronucleus and transgenic rodent gene mutation tests to detect rodent carcinogens.Mutat. Res. 802, 1-29 (2016)
  117. Ahlberg E, Amberg A, Beilke LD, Bower D, Cross KP, Custer L, Ford KA, Van Gompel J, Harvey J, Honma M, Jolly R, Joossens E, Kemper RA, Kenyon M, Kruhlak N, Kuhnke L, Leavitt P, Naven R, Neilan C, Quigley DP, Shuey D, Spirkl HP, Stavitskaya L, Teasdale A, White A, Wichard J, Zwickl C, Myatt GJ. Extending (Q)SARs to incorporate proprietary knowledge for regulatory purposes: A case study using aromatic amine mutagenicity. Regul Toxicol Pharmacol. 13, 1-12 (2016)
  118. Amberg A, Beilke L, Bercu J, Bower D, Brigo A, Cross KP, Custer L, Dobo K, Dowdy E, Ford KA, Glowienke S, Van Gompel J, Harvey J, Hasselgren C, Honma M, Jolly R, Kemper R, Kenyon M, Kruhlak N, Leavitt P, Miller S, Muster W, Nicolette J, Plaper A, Powley M, Quigley DP, Reddy MV, Spirkl HP, Stavitskaya L, Teasdale A, Weiner S, Welch DS, White A, Wichard J, Myatt GJ. Principles and procedures for implementation of ICH M7 recommended (Q)SAR analyses. Regul Toxicol Pharmacol. 77, 13-24 (2016)
  119. Sugiyama K, Yamada M, Awogi T, Hakura A, The strains recommended for use in the bacterial reverse mutation test (OECD guideline 471) can be certified as non-genetically modified organisms. Genes and Environment. 38:2 (2016).
  120. Honma, M., Evaluation of the in vivo genotoxicity of Allura Red AC (Food Red No. 40).Food and Chemical Toxicology. 84, 270-275 (2015).
  121. 本間正充:第1章 ICH M7の適用範囲と既存ガイドラインとの関連性 「ICH M7 変異原性不純物の開発段階からの評価・管理と申請/CTD記載法」サイエンステクノロジー (2015)
  122. 本間正充:1.3遺伝毒性試験法「衛生試験法・注解」金原出版格式会社(日本薬学会編)(2015)
  123. Masumura K, Sakamoto Y, Kumita W, Honma M, Nishikawa A, Nohmi T, Genomic integration of lambda EG10 transgene in gpt delta transgenic rodents. Genes and Environment 37:24 (2015).
  124. Sassa A, Kamoshita N, Kanemaru Y, Honma M, Yasui M, Xeroderma Pigmentosum Group A Suppresses Mutagenesis Caused by Clustered Oxidative DNA Adducts in the Human Genome. PLoS One. 10(11):e0142218 (2015).
  125. Barber C, Amberg A, Custer L, Dobo KL, Glowienke S, Van Gompel J, Gutsell S, Harvey J, Honma M, Kenyon MO, Kruhlak N, Muster W, Stavitskaya L, Teasdale A, Vessey J, Wichard J., Establishing best practise in the application of expert review of mutagenicity under ICH M7. Regul Toxicol Pharmacol. 73, 367-377 (2015)
  126. Canipa S, Cayley A, Drewe WC, Williams RV, Hamada S, Hirose A, Honma M, Morita T. Using in vitro structural alerts for chromosome damage to predict in vivo activity and direct future testing. Mutagenesis 31, 17-25 (2015)
  127. Kanemaru Y, Suzuki T, Niimi N, Grúz P, Matsumoto K, Adachi N, Honma M, Nohmi T. Catalytic and non-catalytic roles of DNA polymerase κ in the protection of human cells against genotoxic stresses. Environ Mol Mutagen. ;56:650-62(2015)
  128. Keka IS, Mohiuddin, Maede Y, Rahman MM, Sakuma T, Honma M, Yamamoto T, Takeda S, Sasanuma H. Smarcal1 promotes double-strand-break repair by nonhomologous end-joining. Nucleic Acids Res. 43, 6359-72 (2015)
  129. Uno Y, Kojima H, Omori T, Corvi R, Honma M, Schechtman LM, Tice RR, Beevers C, Boeck MD, Burlinson B, Hobbs CA, Kitamoto S, Kraynak AR, McNamee J, Nakagawa Y, Pant K, Plappert-Helbig U, Priestley C, Takasawa H, Wada K, Wirnitzer U, Asano N, Escobar PA, Lovell D, Morita T, Nakajima M, Ohno Y, Hayashi M. JaCVAM-organized international validation study of the in vivo rodent alkaline comet assay for detection of genotoxic carcinogens: II. Summary of definitive validation study results. Mutat Res Genet Toxicol Environ Mutagen. 786-788:45-76 (2015)
  130. Morita T, Uno Y, Honma M, Kojima H, Hayashi M, Tice RR, Corvi R, Schechtman L. The JaCVAM international validation study on the in vivo comet assay: Selection of test chemicals. Mutat Res Genet Toxicol Environ Mutagen. 786-788, 14-44 (2015)
  131. Uno Y, Kojima H, Omori T, Corvi R, Honma M, Schechtman LM, Tice RR, Burlinson B, Escobar PA, Kraynak AR, Nakagawa Y, Nakajima M, Pant K, Asano N, Lovell D, Morita T, Ohno Y, Hayashi M. JaCVAM-organized international validation study of the in vivo rodent alkaline comet assay for the detection of genotoxic carcinogens: I. Summary of pre-validation study results. Mutat Res Genet Toxicol Environ Mutagen. 786-788:3-13 (2015)
  132. Hamada S, Ohyama W, Takashima R, Shimada K, Matsumoto K, Kawakami S, Uno F, Sui H, Shimada Y, Imamura T, Matsumura S, Sanada H, Inoue K, Muto S, Ogawa I, Hayashi A, Takayanagi T, Ogiwara Y, Maeda A, Okada E, Terashima Y, Takasawa H, Narumi K, Wako Y, Kawasako K, Sano M, Ohashi N, Morita T, Kojima H, Honma M, Hayashi M. Evaluation of the repeated-dose liver and gastrointestinal tract micronucleus assays with 22 chemicals using young adult rats: summary of the collaborative study by the Collaborative Study Group for the Micronucleus Test (CSGMT)/The Japanese Environmental Mutagen Society (JEMS) – Mammalian Mutagenicity Study Group (MMS). Mutat Res Genet Toxicol Environ Mutagen. 780-781:2-17 (2015)
  133. Martus HJ, Hayashi M, Honma M, Kasper P, Gollapudi B, Mueller L, Schoeny R, Uno Y, Kirkland DJ, Summary of major conclusions from the 6th International Workshop on Genotoxicity Testing (IWGT), Foz do Iguacu, Brazil, Mutat. Res., 783, 1-5 (2015)
  134. Yauk CL, Aardema MJ, Benthem VJ, Bishop JB, Dearfield KL, DeMarini DM, Dubrova YE, Honma M, Lupski JR, Marchetti F, Meistrich ML, Pacchierotti F, Stewart J, Waters MD, Douglas GR. Approaches for Identifying Germ Cell Mutagens: Report of the 2013 IWGT Workshop on Germ Cell Assays. Mutat. Res., 783, 36-54 (2015)
  135. Petkov PI, Patlewicz G, Schultz TW, Honma M, Todorov M, Kotov S, Dimitrov SD, Donner EM, Mekenyan OG, A feasibility study: Can information collected to classify for mutagenicity be informative in predicting carcinogenicity? Regul. Tox. Pharm., 72, 17-25 (2015)
  136. Zeiger E, Gollapudi B, Aardema MJ, Auerbach S, Boverhof D, Custer L, Dedon P, Honma M, Ishida S, Kasinski AL, Kim JH, Manjanatha MG, Marlowe J, Pfuhler S, Pogribny I, Slikker W, Stankowski LF Jr, Tanir JY, Tice R, van Benthem J, White P, Witt KL, Thybaud V. Opportunities to integrate new approaches in genetic toxicology: an ILSI-HESI workshop report. Environ Mol Mutagen. 56, 277-85 (2015)
  137. Aoki Y, Akiko A, Sugawara Y, Hiyoshi-Arai K, Goto S, Masumura K, Nohmi T, Alterations in the mutagenicity and mutation spectrum induced by benzo[a]pyrene instilled in the lungs of gpt delta mice of various ages, Genes and Environment, 37:7 (2015)
  138. Kuroda K, Hibi D, Ishii Y, Yokoo Y, Takasu S, Kijima A, Matsushita K, Masumura K, Kodama Y, Yanai T, Sakai H, Nohmi T, Ogawa K, Umemura T, Role of p53 in the progression from ochratoxin A-induced DNA damage to gene mutations in the kidneys of mice, Toxicol Sci, 144, 65-76 (2015)
  139. Gollapudi BB, Lynch AM, Heflich RH, Dertinger SD, Dobrovolsky VN, Froetschl R, Horibata K, Kenyon MO, Kimoto T, Lovell DP, Stankowski LF Jr, White PA, Witt KL, Tanir JY, The in vivo Pig-a assay: A report of the International Workshop On Genotoxicity Testing (IWGT) Workgroup, Mutat Res Genet Toxicol Environ Mutagen, 1, 23-35 (2015)
  140. Horibata K, Kono S, Ishigami C, Zhang X, Aizawa M, Kako Y, Ishii T, Kosaki R, Saijo M, Tanaka K, Constructive rescue of TFIIH instability by an alternative isoform of XPD derived from a mutated XPD allele in mild but not severe XP-D/CS, J Hum Genet, 60, 259-65 (2015)
  141. Matsuda T, Matsuda S, Yamada M, Mutation assay using single-molecule real-time (SMRTTM) sequencing technology, Genes and Environment, 37: 15 (2015)
  142. Yasui M, Kamoshita N, Nishimura T, Honma M, Mechanism of induction of binucleated cells by multiwalled carbon nanotubes as revealed by live-cell imaging analysis, Genes and Environment, 37: 6 (2015)
  143. Sugiyama K, Takamune M, Furusawa H, Honma M, Human DNA methyltransferase gene-transformed yeasts display an inducible flocculation inhibited by 5-aza-2'-deoxycytidine, Biochem Biophys Res Commun, 456, 689-694 (2015)
  144. Wakasugi M, Sasaki T, Matsumoto M, Nagaoka M, Inoue K, Inobe M, Horibata K, Tanaka K, Matsunaga T, Nucleotide excision repair-dependent DNA double-strand break formation and ATM signaling activation in mammalian quiescent cells, J Biol Chem, 289, 28730-7 (2014)
  145. Onami S, Cho YM, Toyoda T, Horibata K, Ishii Y, Umemura T, Honma M, Nohmi T, Nishikawa A, Ogawa K, Absence of in vivo genotoxicity of 3-monochloropropane-1,2-diol and associated fatty acid esters in a 4-week comprehensive toxicity study using F344 gpt delta rats, Mutagenesis, 29, 295-302 (2014)
  146. Matsumoto, M., Masumori, S., Hirata-Koizumi, M., Ono, A., Honma, M., Yokoyama, K. and Hirose, A., Evaluation of in vivo mutagenicity of hydroquinone in Muta™ mice. Mutat Res Genet Toxicol Environ Mutagen., 775-776, 94-98 (2014)
  147. Morita, T., Miyajima, A., Hatano, A., Honma, M.: Effects of the proposed top concentration limit on an in vitro chromosomal aberration test to assay sensitivity or to reduce the number of false positives, Mutation Research, 769, 34–49 (2014)
  148. Takeiri A, Wada NA, Motoyama S, Matsuzaki K, Tateishi H, Matsumoto K, Niimi N, Sassa A, Grúz P, Masumura K, Yamada M, Mishima M, Jishage K, Nohmi T, In vivo evidence that DNA polymerase kappa is responsible for error-free bypass across DNA cross-links induced by mitomycin C, DNA Repair, 24, 113-121 (2014)
  149. Horibata K, Ukai A, Honma M, Evaluation of rats' in vivo genotoxicity induced by N-ethyl-N-nitrosourea in the RBC Pig-a, PIGRET, and gpt assays, Genes and Enviromment, 36, 199-202 (2014)
  150. Sassa A, Suzuki T, Kanemaru Y, Niimi N, Fujimoto H, Katafuchi A, Grúz P, Yasui M, Gupta RC, Johnson F, Ohta T, Honma M, Adachi N, Nohmi T, In vivo evidence that phenylalanine 171 acts as a molecular brake for translesion DNA synthesis across benzo[a]pyrene DNA adducts by human DNA polymerase κ, DNA Repair, 15, 21-28 (2014)
  151. Yasui M, Kanemaru Y, Kamoshita N, Suzuki T, Arakawa T, Honma M, Tracing the fates of site-specifically introduced DNA adducts in the human genome, DNA Repair, 15, 11-20 (2014)
  152. Grúz P, Sassa A, Hosoda A, Yamagishi H, Usui Y, Shimizu M, Exclusive induction of G:C to A:T transitions by 3-azido-1,2-propanediol in yeast, Mutation Res, 760, 73-76 (2014)
  153. Kuroda K, Hibi D, Ishii Y, Takasu S, Kijima A, Matsushita K, Masumura K, Watanabe M, Sugita-Konishi Y, Sakai H, Yanai T, Nohmi T, Ogawa K, Umemura T, Ochratoxin A induces DNA double-strand breaks and large deletion mutations in the carcinogenic target site of gpt delta rats, Mutagenesis, 29, 27-36 (2014)
  154. Horibata K, Ukai A, Kimoto T, Suzuki T, Kamoshita N, Masumura K, Nohmi T, Honma M, Evaluation of in vivo genotoxicity induced by N-ethyl-N-nitrosourea, benzo[a]pyrene, and 4-nitroquinoline-1-oxide in the Pig-a and gpt assays, Environ Mol Mutagen, 54, 747-754 (2013)
  155. Kawamura Y, Hayashi H, Kurata Y, Hiratsuka K, Masumura K, Nohmi T, Evaluation of the genotoxicity of tamoxifen in the liver and kidney of F344 gpt delta transgenic rat in 3-week and 13-week repeated dose studies, Toxicology, 312, 56-62 (2013)
  156. Sugiyama K, Yamazaki R, Kinoshita M, Kamata Y, Tani F, Minai Y, Sugita-Konishi Y, Inhibitory effect of citrinin on lipopolisaccharide-induced nitric oxide production by mouse macrophage cells, Mycotoxin Res, 29, 229-234 (2013)
  157. Kimura A, Miyata A, Honma M, A combination of in vitro comet assay and micronucleus test using human lymphoblastoid TK6 cells, Mutagenesis, 28, 583-590 (2013)
  158. Shah N, Montes de Oca M, Jover-Cobos M, Tanamoto K, Muroi M, Sugiyama K, Davies NA, Mookerjee RP, Dhar DK, Jalan R, Role of toll-like receptor 4 in mediating multiorgan dysfunction in mice with acetaminophen induced acute liver failure, Liver Transpl, 19, 751-761 (2013)
  159. Kimoto T, Horibata K, Chikura S, Hashimoto K, Itoh S, Sanada H, Muto S, Uno Y, Yamada M, Honma M, Interlaboratory trial of the rat Pig-a mutation assay using an erythroid marker HIS49 antibody, Mutat Res, 755, 126-134 (2013)
  160. Bétous R, Pillaire MJ, Pierini L, van der Laan S, Recolin B, Ohl-Séguy E, Guo C, Niimi N, Grúz P, Nohmi T, Friedberg E, Cazaux C, Maiorano D, Hoffmann JS, DNA polymerase κ-dependent DNA synthesis at stalled replication forks is important for CHK1 activation, EMBO J, 32, 2172-2185 (2013)
  161. Pfuhler S, Elespuru R, Aardema MJ, Doak SH, Maria Donner E, Honma M, Kirsch-Volders M, Landsiedel R, Manjanatha M, Singer T, Kim JH, Genotoxicity of nanomaterials: refining strategies and tests for hazard identification, Environ Mol Mutagen, 54, 229-239 (2013)
  162. T. Matsuda, M. Takamune, Y. Matsuda, M. Yamada, A pilot study for the mutation assay using a high-throughput DNA sequencer, Genes and Environment 35, 53-56 (2013)
  163. P. Grúz, T. Nohmi, Expression and activity of human DNA polymerase η in Escherichia coli, Genes and Environment 35, 10-20 (2013)
  164. A. Sassa, N. Kamoshita, T. Matsuda, Y. Ishii, I. Kuraoka, T. Nohmi, T. Ohta, M. Honma, M. Yasui, Miscoding properties of 8-chloro-2'-deoxyguanosine, a hypochlorous acid-induced DNA adduct, catalysed by human DNA polymerases, Mutagenesis 28, 81-88 (2013)
  165. M. Yamada, M. Shimizu, A. Katafuchi, P. Grúz, S. Fujii, Y. Usui, R.P. Fuchs, T. Nohmi, Escherichia coli DNA polymerase III is responsible for the high level of spontaneous mutations in mutT strains, Mol Microbiol, 86, 1364-75 (2012)
  166. M. Honma, T. Takahashi, S. Asada, Y. Nakagawa, A. Ikeda, K. Yamakage, In vitro clastogenicity and phototoxicity of fullerene (C(60)) nanomaterials in mammalian cells, Mutat Res. 749, 97-100 (2012)
  167. T. Nohmi, M. Honma, M. Yamada, K. Masumura, M. Yasui, K. Horibata, S. Fukushima, 2nd International Symposium on Genotoxic and Carcinogenic Thresholds, Genes and Environment 34, 141-145 (2012)
  168. T. Nohmi, M. Yamada, K. Masumura, In vivo Approaches to Identify Mutations and in vitro Research to Reveal Underlying Mechanisms of Genotoxic Thresholds, Genes and Environment 34, 146-152 (2012)
  169. A.D. Bailey, L.T. Gray, T. Pavelitz, J.C. Newman, K. Horibata, K. Tanaka, A.M. Weiner, The conserved Cockayne syndrome B-piggyBac fusion protein (CSB-PGBD3) affects DNA repair and induces both interferon-like and innate antiviral responses in CSB-null cells, DNA repair, 11, 488-501 (2012)
  170. X. Zhang, K. Horibata, M. Saijo, C. Ishigami, A. Ukai, S.I. Kanno, H. Tahara, E.G. Neilan, M. Honma, T. Nohmi, A. Yasui, K. Tanaka, Mutations in UVSSA cause UV-sensitive syndrome and destabilize ERCC6 in transcription-coupled DNA repair, Nature Genetics, 44, 593-597 (2012)
  171. G. Xing, X. Qia, M. Chen, Y. Wu, J. Yao, L. Gong, T. Nohmi, Y. Luan, J. Ren, Comparison of the mutagenicity of aristolochic acid I and aristolochic acid II in the gpt delta transgenic mouse kidney, Mutat Res, 743, 52–58 (2012)
  172. O.G. Mekenyan, P.I. Petkov, S.V. Kotov, S. Stoeva, V.B. Kamenska, S.D. Dimitrov, M. Honma, M. Hayashi, R. Benigni, E.M. Donner, G. Patlewicz, Investigating the relationship between in vitro-in vivo genotoxicity: Derivation of mechanistic QSAR models for in vivo liver genotoxicity and in vivo bone marrow micronucleus formation which encompass metabolism, Chem Res Toxicol, 25, 277-96 (2012)
  173. F. Yatagai, M. Honma, A. Ukai, K. Ohmori, H. Suzuki, T. Shimizu, A. Takahashi, T. Ohnishi, N. Dohmae, N. Ishioka, Preliminary results of space experiment: Implications for the effects of space radiation and microgravity on survival and mutation induction in human cells, Advance in Space Research 49, 479-485 (2012)
  174. T. Kamigaito, T. Noguchi, K. Narumi, R. Takashima, S. Hamada, H. Sanada, M. Hasuko, H. Hayashi, K. Masumura, T. Nohmi, Evaluation of the in vivo mutagenicity of nickel subsulfide in the lung of F344 gpt delta transgenic rats exposed by intratracheal instillation: A collaborative study for the gpt delta transgenic rat mutation assay, Genes and Environment, 34, 34-44 (2012)
  175. H. Sui, R. Ohta, T. Shiragiku, A. Akahori, K. Suzuki, M. Nakajima, H. Hayashi, K. Masumura, T. Nohmi, Evaluation of in vivo mutagenicity by 2,4-diaminotoluene and 2,6-diaminotoluene in liver of F344 gpt delta transgenic rat dosed for 28 days: A collaborative study of the gpt delta transgenic rat mutation assay, Genes and Environment, 34, 25-33 (2012)
  176. Y. Kawamura, H. Hayashi, O. Tajima, S. Yamada, T. Takayanagi, H. Hori, W. Fujii, K. Masumura, T. Nohmi, Evaluation of the genotoxicity of aristolochic acid in the kidney and liver of F344 gpt delta transgenic rat using a 28-day repeated-dose protocol: A collaborative study of the gpt delta transgenic rat mutation assay, Genes and Environment, 34, 18-24 (2012)

2012年以前の研究業績

ページトップへ