User menu

Accès à distance ? S'identifier sur le proxy UCLouvain

Genotoxicity of engineered nanomaterials: A critical review

  1. Albertini Richard J, Anderson Diana, Douglas George R, Hagmar Lars, Hemminki Kari, Merlo Franco, Natarajan A.T, Norppa Hannu, Shuker David E.G, Tice Raymond, Waters Michael D, Aitio Antero, IPCS guidelines for the monitoring of genotoxic effects of carcinogens in humans, 10.1016/s1383-5742(00)00049-1
  2. Asmuss M., Differential effects of toxic metal compounds on the activities of Fpg and XPA, two zinc finger proteins involved in DNA repair, 10.1093/carcin/21.11.2097
  3. Asmuß Monika, Mullenders Leon H.F., Hartwig Andrea, Interference by toxic metal compounds with isolated zinc finger DNA repair proteins, 10.1016/s0378-4274(99)00273-8
  4. Auffan Melanie, Decome Laetitia, Rose Jerome, Orsiere Thierry, De Meo Michel, Briois Valerie, Chaneac Corinne, Olivi Luca, Berge-lefranc Jean-louis, Botta Alain, Wiesner Mark R., Bottero Jean-yves, In Vitro Interactions between DMSA-Coated Maghemite Nanoparticles and Human Fibroblasts:  A Physicochemical and Cyto-Genotoxical Study†, 10.1021/es060691k
  5. Belyanskaya Larisa, Manser Pius, Spohn Philipp, Bruinink Arie, Wick Peter, The reliability and limits of the MTT reduction assay for carbon nanotubes–cell interaction, 10.1016/j.carbon.2007.08.010
  6. Brown D M, Increased inflammation and intracellular calcium caused by ultrafine carbon black is independent of transition metals or other soluble components, 10.1136/oem.57.10.685
  7. Brown D.M., Wilson M.R., MacNee W., Stone V., Donaldson K., Size-Dependent Proinflammatory Effects of Ultrafine Polystyrene Particles: A Role for Surface Area and Oxidative Stress in the Enhanced Activity of Ultrafines, 10.1006/taap.2001.9240
  8. Cammerer Z., Elhajouji A., Kirsch-Volders M., Suter W., Comparison of the peripheral blood micronucleus test using flow cytometry in rat and mouse exposed to aneugens after single-dose applications, 10.1093/mutage/gel066
  9. Cammerer Zoryana, Elhajouji Azeddine, Suter Willi, In vivo micronucleus test with flow cytometry after acute and chronic exposures of rats to chemicals, 10.1016/j.mrgentox.2006.08.004
  10. Casey A., Herzog E., Davoren M., Lyng F.M., Byrne H.J., Chambers G., Spectroscopic analysis confirms the interactions between single walled carbon nanotubes and various dyes commonly used to assess cytotoxicity, 10.1016/j.carbon.2007.03.033
  11. Chae Sunyoung, Yun Chawon, Um Haeryun, Lee Jae-Ho, Cho Hyeseong, Centrosome amplification and multinuclear phenotypes are Induced by hydrogen peroxide, 10.1038/emm.2005.59
  12. Chanda Sarmishtha, Dasgupta Uma B., GuhaMazumder Debendranath, Gupta Mausumi, Chaudhuri Utpal, Lahiri Sarbari, Das Subhankar, Ghosh Nilima, Chatterjee Debdutta, DNA Hypermethylation of Promoter of Gene p53 and p16 in Arsenic-Exposed People with and without Malignancy, 10.1093/toxsci/kfj030
  13. CHEN M, VONMIKECZ A, Formation of nucleoplasmic protein aggregates impairs nuclear function in response to SiO nanoparticles, 10.1016/j.yexcr.2004.12.021
  14. Chen H., Chronic inorganic arsenic exposure induces hepatic global and individual gene hypomethylation: implications for arsenic hepatocarcinogenesis, 10.1093/carcin/bgh161
  15. Conlon Kimberly A., Miller Holly, Rosenquist Thomas A., Zharkov Dmitry O., Berrios Miguel, The murine DNA glycosylase NEIL2 (mNEIL2) and human DNA polymerase β bind microtubules in situ and in vitro, 10.1016/j.dnarep.2004.10.010
  16. Conlon Kimberly A., Zharkov Dmitry O., Berrios Miguel, Cell cycle regulation of the murine 8-oxoguanine DNA glycosylase (mOGG1): mOGG1 associates with microtubules during interphase and mitosis, 10.1016/j.dnarep.2004.06.011
  17. Coyle P., Philcox J. C., Carey L. C., Rofe A. M., Metallothionein: the multipurpose protein, 10.1007/s00018-002-8454-2
  18. Daniel LN, Scand J Work Environ Health (Suppl.)2, 21, 22 (1995)
  19. De Boeck M., In vivo genotoxicity of hard metal dust: induction of micronuclei in rat type II epithelial lung cells, 10.1093/carcin/bgg146
  20. De Boeck M, Evaluation of the in vitro direct and indirect genotoxic effects of cobalt compounds using the alkaline comet assay. Influence of interdonor and interexperimental variability, 10.1093/carcin/19.11.2021
  21. Decordier Ilse, Cundari Enrico, Kirsch-Volders Micheline, Mitotic checkpoints and the maintenance of the chromosome karyotype, 10.1016/j.mrgentox.2007.10.020
  22. Decordier Ilse, De Bont Kelly, De Bock Kirsten, Mateuca Raluca, Roelants Mathieu, Ciardelli Roberta, Haumont Dominique, Knudsen Lisbeth E., Kirsch-Volders Micheline, Genetic susceptibility of newborn daughters to oxidative stress, 10.1016/j.toxlet.2007.05.014
  23. Driscoll K., Effects of particle exposure and particle-elicited inflammatory cells on mutation in rat alveolar epithelial cells, 10.1093/carcin/18.2.423
  24. Elhajouji Azeddine, Tibaldi Fabian, Kirsch-Volders Micheline, Indication for thresholds of chromosome non-disjunction versus chromosome lagging induced by spindle inhibitors in vitro in human lymphocytes, 10.1093/mutage/12.3.133
  25. Elhajouji Azeddine, Van Hummelen Paul, Kirsch-Volders Micheline, Indications for a threshold of chemically-induced aneuploidy in vitro in human lymphocytes, 10.1002/em.2850260405
  26. Fenech Michael, The in vitro micronucleus technique, 10.1016/s0027-5107(00)00065-8
  27. Fenech Michael, Cytokinesis-block micronucleus cytome assay, 10.1038/nprot.2007.77
  28. Fenoglio Ivana, Tomatis Maura, Lison Dominique, Muller Julie, Fonseca Antonio, Nagy Janos B., Fubini Bice, Reactivity of carbon nanotubes: Free radical generation or scavenging activity?, 10.1016/j.freeradbiomed.2005.11.010
  29. Fubini Bice, Hubbard Andrea, Reactive oxygen species (ROS) and reactive nitrogen species (RNS) generation by silica in inflammation and fibrosis, 10.1016/s0891-5849(03)00149-7
  30. Garcia VAP, Eur Cells Materials, 3, 154 (2002)
  31. Geiser Marianne, Rothen-Rutishauser Barbara, Kapp Nadine, Schürch Samuel, Kreyling Wolfgang, Schulz Holger, Semmler Manuela, Hof Vinzenz Im, Heyder Joachim, Gehr Peter, Ultrafine Particles Cross Cellular Membranes by Nonphagocytic Mechanisms in Lungs and in Cultured Cells, 10.1289/ehp.8006
  32. GILLI G, TRAVERSI D, ROVERE R, PIGNATA C, SCHILIRO T, Chemical characteristics and mutagenic activity of PM10 in Torino, a Northern Italian City, 10.1016/j.scitotenv.2007.07.006
  33. Gurr Jia-Ran, Wang Alexander S.S., Chen Chien-Hung, Jan Kun-Yan, Ultrafine titanium dioxide particles in the absence of photoactivation can induce oxidative damage to human bronchial epithelial cells, 10.1016/j.tox.2005.05.007
  34. Hagmar Lars, Strömberg Ulf, Bonassi Stefano, Hansteen Inger-Lise, Knudsen Lisbeth Ehlert, Lindholm Carita, Norppa Hannu, Impact of Types of Lymphocyte Chromosomal Aberrations on Human Cancer Risk : Results from Nordic and Italian Cohorts, 10.1158/0008-5472.can-03-3360
  35. Hartwig A, In t J Cancer, 104, 1 (2003)
  36. Harush-Frenkel Oshrat, Debotton Nir, Benita Simon, Altschuler Yoram, Targeting of nanoparticles to the clathrin-mediated endocytic pathway, 10.1016/j.bbrc.2006.11.135
  37. Huang Min, Ma Zengshuan, Khor Eugene, Lim Lee‐Yong, 10.1023/a:1020404615898
  38. Huang S., Arsenite inhibits mitotic division and perturbs spindle dynamics in HeLa S3 cells, 10.1093/carcin/19.5.889
  39. Jacobsen Nicklas Raun, Saber Anne Thoustrup, White Paul, Møller Peter, Pojana Giulio, Vogel Ulla, Loft Steffen, Gingerich John, Soper Lynda, Douglas George R., Wallin Håkan, Increased mutant frequency by carbon black, but not quartz, in thelacZ andcII transgenes of muta™mouse lung epithelial cells, 10.1002/em.20300
  40. Jaurand MC, IARC Sci Publ, 90, 54 (1989)
  41. Jin Yuhui, Kannan Shibichakravarthy, Wu Min, Zhao Julia Xiaojun, Toxicity of Luminescent Silica Nanoparticles to Living Cells, 10.1021/tx7001959
  42. Kasten Ursula, Mullenders Leon H.F, Hartwig Andrea, Cobalt(II) inhibits the incision and the polymerization step of nucleotide excision repair in human fibroblasts, 10.1016/s0921-8777(96)00052-3
  43. Kim Jun Sung, Yoon Tae-Jong, Yu Kyeong Nam, Kim Byung Gul, Park Sung Jin, Kim Hyun Woo, Lee Kee Ho, Park Seung Bum, Lee Jin-Kyu, Cho Myung Haing, Toxicity and Tissue Distribution of Magnetic Nanoparticles in Mice, 10.1093/toxsci/kfj027
  44. Kim Jun-Sung, Yoon Tae-Jong, Yu Kyeong-Nam, Noh Mi Suk, Woo Minah, Kim Byung-Geol, Lee Kee-Ho, Sohn Byung-Hyuk, Park Seung-Bum, Lee Jin-Kyu, Cho Myung-Haing, Cellular uptake of magnetic nanoparticle is mediated through energy-dependent endocytosis in A549 cells, 10.4142/jvs.2006.7.4.321
  45. Kirsch-Volders Micheline, Aardema Marilyn, Elhajouji Azeddine, Concepts of threshold in mutagenesis and carcinogenesis, 10.1016/s1383-5718(99)00161-8
  46. Kirsch-Volders M., Inclusion of micronuclei in non-divided mononuclear lymphocytes and necrosis/apoptosis may provide a more comprehensive cytokinesis block micronucleus assay for biomonitoring purposes, 10.1093/mutage/16.1.51
  47. Kirsch-Volders Micheline, Vanhauwaert Annelies, De Boeck Marlies, Decordier Ilse, Importance of detecting numerical versus structural chromosome aberrations, 10.1016/s0027-5107(02)00087-8
  48. Kirsch-Volders Micheline, Vanhauwaert Annelies, Eichenlaub-Ritter Ursula, Decordier Ilse, Indirect mechanisms of genotoxicity, 10.1016/s0378-4274(02)00498-8
  49. Knaapen Ad M., Borm Paul J.A., Albrecht Catrin, Schins Roel P.F., Inhaled particles and lung cancer. Part A: Mechanisms, 10.1002/ijc.11708
  50. Knaapen Ad M., Shi Tingming, Borm Paul J.A., Schins Roel P.F., 10.1023/a:1015970023889
  51. Lewinski Nastassja, Colvin Vicki, Drezek Rebekah, Cytotoxicity of Nanoparticles, 10.1002/smll.200700595
  52. Limbach Ludwig K., Wick Peter, Manser Pius, Grass Robert N., Bruinink Arie, Stark Wendelin J., Exposure of Engineered Nanoparticles to Human Lung Epithelial Cells:  Influence of Chemical Composition and Catalytic Activity on Oxidative Stress, 10.1021/es062629t
  53. Lison Dominique, Carbonnelle Philippe, Mollo Laura, Lauwerys Robert, Fubini Bice, Physicochemical Mechanism of the Interaction between Cobalt Metal and Carbide Particles To Generate Toxic Activated Oxygen Species, 10.1021/tx00046a015
  54. MAENOSONO Shinya, SUZUKI Toshimasa, SAITA Soichiro, MUTAGENICITY OF WATER-SOLUBLE FePt NANOPARTICLES IN AMES TEST, 10.2131/jts.32.575
  55. Marnett Lawrence J., Oxyradicals and DNA damage, 10.1093/carcin/21.3.361
  56. Maxwell Patrick, Salnikow Konstantin, HIF-1, An Oxygen and Metal Responsive Transcription Factor, 10.4161/cbt.3.1.547
  57. Möller Winfried, Brown David M, Kreyling Wolfgang G, Stone Vicki, 10.1186/1743-8977-2-7
  58. Monteiro-Riviere Nancy A., Inman Alfred O., Challenges for assessing carbon nanomaterial toxicity to the skin, 10.1016/j.carbon.2005.11.004
  59. Mroz R. M., Schins R. P. F., Li H., Jimenez L. A., Drost E. M., Holownia A., MacNee W., Donaldson K., Nanoparticle-driven DNA damage mimics irradiation-related carcinogenesis pathways, 10.1183/09031936.00006707
  60. Muller Julie, Decordier Ilse, Hoet Peter H., Lombaert Noömi, Thomassen Leen, Huaux François, Lison Dominique, Kirsch-Volders Micheline, Clastogenic and aneugenic effects of multi-wall carbon nanotubes in epithelial cells, 10.1093/carcin/bgm243
  61. Murdock Richard C., Braydich-Stolle Laura, Schrand Amanda M., Schlager John J., Hussain Saber M., Characterization of Nanomaterial Dispersion in Solution Prior to In Vitro Exposure Using Dynamic Light Scattering Technique, 10.1093/toxsci/kfm240
  62. Nabiev Igor, Mitchell Siobhan, Davies Anthony, Williams Yvonne, Kelleher Dermot, Moore Richard, Gun'ko Yurii K., Byrne Stephen, Rakovich Yury P., Donegan John F., Sukhanova Alyona, Conroy Jennifer, Cottell David, Gaponik Nikolai, Rogach Andrey, Volkov Yuri, Nonfunctionalized Nanocrystals Can Exploit a Cell's Active Transport Machinery Delivering Them to Specific Nuclear and Cytoplasmic Compartments, 10.1021/nl0719832
  63. Niwa Yasuharu, Iwai Naoharu, Genotoxicity in cell lines induced by chronic exposure to water-soluble fullerenes using micronucleus test, 10.1007/bf02898019
  64. Oberdörster G, Ferin J, Gelein R, Soderholm S C, Finkelstein J, Role of the alveolar macrophage in lung injury: studies with ultrafine particles, 10.1289/ehp.9297193
  65. Oberdörster Günter, Maynard Andrew, Donaldson Ken, Castranova Vincent, Fitzpatrick Julie, Ausman Kevin, Carter Janet, Karn Barbara, Kreyling Wolfgang, Lai David, Olin Stephen, Monteiro-Riviere Nancy, Warheit David, Yang Hong, 10.1186/1743-8977-2-8
  66. Oberdörster Günter, Oberdörster Eva, Oberdörster Jan, Nanotoxicology: An Emerging Discipline Evolving from Studies of Ultrafine Particles, 10.1289/ehp.7339
  67. Pante N., Nuclear Pore Complex Is Able to Transport Macromolecules with Diameters of 39 nm, 10.1091/mbc.01-06-0308
  68. Panyam Jayanth, Labhasetwar Vinod, 10.1023/a:1022219003551
  69. PANYAM J., Rapid endo-lysosomal escape of poly(DL-lactide-co-glycolide) nanoparticles: implications for drug and gene delivery, 10.1096/fj.02-0088com
  70. Papageorgiou I., Brown C., Schins R., Singh S., Newson R., Davis S., Fisher J., Ingham E., Case C.P., The effect of nano- and micron-sized particles of cobalt–chromium alloy on human fibroblasts in vitro, 10.1016/j.biomaterials.2007.02.034
  71. Park Barry, Martin Patricia, Harris Chris, Guest Robert, Whittingham Andrew, Jenkinson Peter, Handley John, Initial in vitro screening approach to investigate the potential health and environmental hazards of Envirox™ – a nanoparticulate cerium oxide diesel fuel additive, 10.1186/1743-8977-4-12
  72. Powers Kevin W., Brown Scott C., Krishna Vijay B., Wasdo Scott C., Moudgil Brij M., Roberts Stephen M., Research Strategies for Safety Evaluation of Nanomaterials. Part VI. Characterization of Nanoscale Particles for Toxicological Evaluation, 10.1093/toxsci/kfj099
  73. PULSKAMP K, DIABATE S, KRUG H, Carbon nanotubes show no sign of acute toxicity but induce intracellular reactive oxygen species in dependence on contaminants, 10.1016/j.toxlet.2006.11.001
  74. Rahman Qamar, Lohani Mohtashim, Dopp Elke, Pemsel Heidemarie, Jonas Ludwig, Weiss Dieter G., Schiffmann Dietmar, Evidence That Ultrafine Titanium Dioxide Induces Micronuclei and Apoptosis in Syrian Hamster Embryo Fibroblasts, 10.1289/ehp.02110797
  75. Reeves James F., Davies Simon J., Dodd Nicholas J.F., Jha Awadhesh N., Hydroxyl radicals (OH) are associated with titanium dioxide (TiO2) nanoparticle-induced cytotoxicity and oxidative DNA damage in fish cells, 10.1016/j.mrfmmm.2007.12.010
  76. Rehn B, Seiler F, Rehn S, Bruch J, Maier M, Investigations on the inflammatory and genotoxic lung effects of two types of titanium dioxide: untreated and surface treated, 10.1016/s0041-008x(03)00092-9
  77. Sadeghiani N., Barbosa L.S., Silva L.P., Azevedo R.B., Morais P.C., Lacava Z.G.M., Genotoxicity and inflammatory investigation in mice treated with magnetite nanoparticles surface coated with polyaspartic acid, 10.1016/j.jmmm.2004.11.131
  78. Schins Roel P. F., MECHANISMS OF GENOTOXICITY OF PARTICLES AND FIBERS, 10.1080/089583701753338631
  79. Schins Roel P. F., Knaapen Ad M., Genotoxicity of Poorly Soluble Particles, 10.1080/08958370701496202
  80. Sevastyanova O., Binkova B., Topinka J., Sram R.J., Kalina I., Popov T., Novakova Z., Farmer P.B., In vitro genotoxicity of PAH mixtures and organic extract from urban air particles, 10.1016/j.mrfmmm.2007.03.002
  81. Shi Qinghua, King Randall W., Chromosome nondisjunction yields tetraploid rather than aneuploid cells in human cell lines, 10.1038/nature03958
  82. Tafazoli M., Kirsch-Volders M., In vitro mutagenicity and genotoxicity study of 1,2-dichloroethylene, 1,1,2-trichloroethane, 1,3-dichloropropane, 1,2,3-trichloropropane and 1,1,3-trichloropropene, using the micronucleus test and the alkaline single cell gel electrophoresis technique (comet assay) in human lymphocytes, 10.1016/s0165-1218(96)90107-x
  83. Theogaraj Elizabeth, Riley Susan, Hughes Laurie, Maier Monika, Kirkland David, An investigation of the photo-clastogenic potential of ultrafine titanium dioxide particles, 10.1016/j.mrgentox.2007.08.002
  84. Tice R. R., Agurell E., Anderson D., Burlinson B., Hartmann A., Kobayashi H., Miyamae Y., Rojas E., Ryu J.-C., Sasaki Y. F., Single cell gel/comet assay: Guidelines for in vitro and in vivo genetic toxicology testing, 10.1002/(sici)1098-2280(2000)35:3<206::aid-em8>3.0.co;2-j
  85. Valko Marian, Izakovic Mario, Mazur Milan, Rhodes Christopher J., Telser Joshua, Role of oxygen radicals in DNA damage and cancer incidence, 10.1023/b:mcbi.0000049134.69131.89
  86. Valko M., Rhodes C.J., Moncol J., Izakovic M., Mazur M., Free radicals, metals and antioxidants in oxidative stress-induced cancer, 10.1016/j.cbi.2005.12.009
  87. Wang David, Kreutzer Deborah A., Essigmann John M., Mutagenicity and repair of oxidative DNA damage: insights from studies using defined lesions, 10.1016/s0027-5107(98)00066-9
  88. Wang Jing J., Sanderson Barbara J.S., Wang He, Cyto- and genotoxicity of ultrafine TiO2 particles in cultured human lymphoblastoid cells, 10.1016/j.mrgentox.2006.12.003
  89. Wang Jing J., Sanderson Barbara J.S., Wang He, Cytotoxicity and genotoxicity of ultrafine crystalline SiO2 particulate in cultured human lymphoblastoid cells, 10.1002/em.20287
  90. Warheit David B., How Meaningful are the Results of Nanotoxicity Studies in the Absence of Adequate Material Characterization?, 10.1093/toxsci/kfm279
  91. WARHEIT D, HOKE R, FINLAY C, DONNER E, REED K, SAYES C, Development of a base set of toxicity tests using ultrafine TiO2 particles as a component of nanoparticle risk management, 10.1016/j.toxlet.2007.04.008
  92. Wörle-Knirsch J. M., Pulskamp K., Krug H. F., Oops They Did It Again! Carbon Nanotubes Hoax Scientists in Viability Assays, 10.1021/nl060177c
  93. Xia Tian, Kovochich Michael, Brant Jonathan, Hotze Matt, Sempf Joan, Oberley Terry, Sioutas Constantinos, Yeh Joanne I., Wiesner Mark R., Nel Andre E., Comparison of the Abilities of Ambient and Manufactured Nanoparticles To Induce Cellular Toxicity According to an Oxidative Stress Paradigm, 10.1021/nl061025k
  94. Zhu Lin, Chang Dong Wook, Dai Liming, Hong Yiling, DNA Damage Induced by Multiwalled Carbon Nanotubes in Mouse Embryonic Stem Cells, 10.1021/nl071303v
Bibliographic reference Gonzalez, Laetitia ; Lison, Dominique ; Kirsch-Volders, Micheline. Genotoxicity of engineered nanomaterials: A critical review. In: Nanotoxicology, Vol. 2, no. 4, p. 252-273 (2008)
Permanent URL http://hdl.handle.net/2078.1/36191