메뉴 건너뛰기




Volumn 35, Issue 2, 2018, Pages 235-253

A High-Throughput Approach to Identify Specific Neurotoxicants / Developmental Toxicants in Human Neuronal Cell Function Assays

Author keywords

cytotoxicity; developmental toxicity; high content imaging; neurite outgrowth inhibition; neurotoxicity

Indexed keywords

CELL CULTURE; DOPAMINERGIC NERVE CELL; DRUG EFFECT; HIGH THROUGHPUT SCREENING; HUMAN; PROCEDURES; TOXICITY TESTING;

EID: 85049303762     PISSN: 1868596X     EISSN: 18688551     Source Type: Journal    
DOI: 10.14573/altex.1712182     Document Type: Article
Times cited : (55)

References (80)
  • 1
    • 85011999800 scopus 로고    scopus 로고
    • Reference compounds for alternative test methods to indicate developmental neurotoxicity (DNT) potential of chemicals: Example lists and criteria for their selection and use
    • Aschner, M., Ceccatelli, S., Daneshian, M. et al. (2017). Reference compounds for alternative test methods to indicate developmental neurotoxicity (DNT) potential of chemicals: Example lists and criteria for their selection and use. ALTEX 34, 49-74. doi:10.14573/altex.1604201
    • (2017) ALTEX , vol.34 , pp. 49-74
    • Aschner, M.1    Ceccatelli, S.2    Daneshian, M.3
  • 2
    • 84922032722 scopus 로고    scopus 로고
    • International stakeholder network (ISTNET): Creating a developmental neurotoxicity (DNT) testing road map for regulatory purposes
    • Bal-Price, A., Crofton, K. M., Leist, M. et al. (2015). International stakeholder network (ISTNET): Creating a developmental neurotoxicity (DNT) testing road map for regulatory purposes. Arch Toxicol 89, 269-287. doi:10.1007/s00204-015-1464-2
    • (2015) Arch Toxicol , vol.89 , pp. 269-287
    • Bal-Price, A.1    Crofton, K. M.2    Leist, M.3
  • 3
    • 84865776716 scopus 로고    scopus 로고
    • Epigenetic changes and disturbed neural development in a human embryonic stem cell-based model relating to the fetal valproate syndrome
    • Balmer, N. V., Weng, M. K., Zimmer, B. et al. (2012). Epigenetic changes and disturbed neural development in a human embryonic stem cell-based model relating to the fetal valproate syndrome. Hum Mol Genet 21, 4104-4114. doi:10.1093/hmg/ dds239
    • (2012) Hum Mol Genet , vol.21 , pp. 4104-4114
    • Balmer, N. V.1    Weng, M. K.2    Zimmer, B.3
  • 5
    • 84937846473 scopus 로고    scopus 로고
    • Comparative human and rat neurospheres reveal species differences in chemical effects on neurodevelopmental key events
    • Baumann, J., Gassmann, K., Masjosthusmann, S. et al. (2016). Comparative human and rat neurospheres reveal species differences in chemical effects on neurodevelopmental key events. Arch Toxicol 90, 1415-1427. doi:10.1007/s00204-015-1568-8
    • (2016) Arch Toxicol , vol.90 , pp. 1415-1427
    • Baumann, J.1    Gassmann, K.2    Masjosthusmann, S.3
  • 6
    • 84977118072 scopus 로고    scopus 로고
    • Project TENDR: Targeting environmental neuro-developmental risks the TENDR consensus statement
    • Bennett, D., Bellinger, D. C., Birnbaum, L. S. et al. (2016). Project TENDR: Targeting environmental neuro-developmental risks the TENDR consensus statement. Environ Health Perspect 124, A118-122. doi:10.1289/ehp358
    • (2016) Environ Health Perspect , vol.124 , pp. A118-A122
    • Bennett, D.1    Bellinger, D. C.2    Birnbaum, L. S.3
  • 7
    • 85018424306 scopus 로고    scopus 로고
    • Assaying spontaneous network activity and cellular viability using multi-well microelectrode arrays
    • Brown, J. P., Lynch, B. S., Curry-Chisolm, I. M. et al. (2017). Assaying spontaneous network activity and cellular viability using multi-well microelectrode arrays. Methods Mol Biol 1601, 153-170. doi:10.1007/978-1-4939-6960-9_13
    • (2017) Methods Mol Biol , vol.1601 , pp. 153-170
    • Brown, J. P.1    Lynch, B. S.2    Curry-Chisolm, I. M.3
  • 8
    • 85172758065 scopus 로고    scopus 로고
    • Experimental and clinical neurotoxicology. Second edition
    • Cavanagh, J. B. (2000). Experimental and clinical neurotoxicology. Second edition. Brain 123, 2571-2573. doi:10.1093/ brain/123.12.2571
    • (2000) Brain , vol.123 , pp. 2571-2573
    • Cavanagh, J. B.1
  • 9
    • 79958270042 scopus 로고    scopus 로고
    • Developmental neurotoxicity testing: Recommendations for developing alternative methods for the screening and prioritization of chemicals
    • Crofton, K. M., Mundy, W. R., Lein, P. J. et al. (2011). Developmental neurotoxicity testing: Recommendations for developing alternative methods for the screening and prioritization of chemicals. ALTEX 28, 9-15. doi:10.14573/altex.2011.1.009
    • (2011) ALTEX , vol.28 , pp. 9-15
    • Crofton, K. M.1    Mundy, W. R.2    Lein, P. J.3
  • 10
    • 84865560583 scopus 로고    scopus 로고
    • Developmental neurotoxicity testing: A path forward
    • Crofton, K. M., Mundy, W. R. and Shafer, T. J. (2012). Developmental neurotoxicity testing: A path forward. Congenit Anom (Kyoto) 52, 140-146. doi:10.1111/j.1741-4520.2012.00377.x
    • (2012) Congenit Anom (Kyoto) , vol.52 , pp. 140-146
    • Crofton, K. M.1    Mundy, W. R.2    Shafer, T. J.3
  • 11
    • 84901820244 scopus 로고    scopus 로고
    • International stakeholder network (ISTNET) for creating a developmental neurotoxicity testing (DNT) roadmap for regulatory purposes
    • Crofton, K., Fritsche, E., Ylikomi, T. et al. (2014). International stakeholder network (ISTNET) for creating a developmental neurotoxicity testing (DNT) roadmap for regulatory purposes. ALTEX 31, 223-224. doi:10.14573/altex.1402121
    • (2014) ALTEX , vol.31 , pp. 223-224
    • Crofton, K.1    Fritsche, E.2    Ylikomi, T.3
  • 12
    • 84871716691 scopus 로고    scopus 로고
    • Comparison of chemical-induced changes in proliferation and apoptosis in human and mouse neuroprogenitor cells
    • Culbreth, M. E., Harrill, J. A., Freudenrich, T. M. et al. (2012). Comparison of chemical-induced changes in proliferation and apoptosis in human and mouse neuroprogenitor cells. Neurotoxicology 33, 1499-1510. doi:10.1016/j.neuro.2012.05.012
    • (2012) Neurotoxicology , vol.33 , pp. 1499-1510
    • Culbreth, M. E.1    Harrill, J. A.2    Freudenrich, T. M.3
  • 13
    • 84961653980 scopus 로고    scopus 로고
    • Highlight report: Launch of a large integrated European in vitro toxicology project: EU-ToxRisk
    • Daneshian, M., Kamp, H., Hengstler, J. et al. (2016). Highlight report: Launch of a large integrated European in vitro toxicology project: EU-ToxRisk. Arch Toxicol 90, 1021-1024. doi:10.1007/s00204-016-1698-7
    • (2016) Arch Toxicol , vol.90 , pp. 1021-1024
    • Daneshian, M.1    Kamp, H.2    Hengstler, J.3
  • 14
    • 70349919668 scopus 로고    scopus 로고
    • Suitability of GRIND-based principal properties for the description of molecular similarity and ligand-based virtual screening
    • Duran, A., Zamora, I. and Pastor, M. (2009). Suitability of GRIND-based principal properties for the description of molecular similarity and ligand-based virtual screening. J Chem Inf Model 49, 2129-2138. doi:10.1021/ci900228x
    • (2009) J Chem Inf Model , vol.49 , pp. 2129-2138
    • Duran, A.1    Zamora, I.2    Pastor, M.3
  • 15
    • 84987645985 scopus 로고    scopus 로고
    • State of the art in non-animal approaches for skin sensitization testing: From individual test methods towards testing strategies
    • Ezendam, J., Braakhuis, H. M. and Vandebriel, R. J. (2016). State of the art in non-animal approaches for skin sensitization testing: From individual test methods towards testing strategies. Arch Toxicol 90, 2861-2883. doi:10.1007/s00204-016-1842-4
    • (2016) Arch Toxicol , vol.90 , pp. 2861-2883
    • Ezendam, J.1    Braakhuis, H. M.2    Vandebriel, R. J.3
  • 16
    • 80855143608 scopus 로고    scopus 로고
    • Effects of sub-lethal neurite outgrowth inhibitory concentrations of chlorpyrifos oxon on cytoskeletal proteins and acetylcholinesterase in differentiating N2a cells
    • Flaskos, J., Nikolaidis, E., Harris, W. et al. (2011). Effects of sub-lethal neurite outgrowth inhibitory concentrations of chlorpyrifos oxon on cytoskeletal proteins and acetylcholinesterase in differentiating N2a cells. Toxicol Appl Pharmacol 256, 330-336. doi:10.1016/j.taap.2011.06.002
    • (2011) Toxicol Appl Pharmacol , vol.256 , pp. 330-336
    • Flaskos, J.1    Nikolaidis, E.2    Harris, W.3
  • 17
    • 77949388450 scopus 로고    scopus 로고
    • The network formation assay: A spatially standardized neurite outgrowth analytical display for neurotoxicity screening
    • Frimat, J. P., Sisnaiske, J., Subbiah, S. et al. (2010). The network formation assay: A spatially standardized neurite outgrowth analytical display for neurotoxicity screening. Lab Chip 10, 701-709. doi:10.1039/b922193j
    • (2010) Lab Chip , vol.10 , pp. 701-709
    • Frimat, J. P.1    Sisnaiske, J.2    Subbiah, S.3
  • 18
    • 22144449590 scopus 로고    scopus 로고
    • Polychlorinated biphenyls disturb differentiation of normal human neural progenitor cells: Clue for involvement of thyroid hormone receptors
    • Fritsche, E., Cline, J. E., Nguyen, N.-H. et al. (2005). Polychlorinated biphenyls disturb differentiation of normal human neural progenitor cells: Clue for involvement of thyroid hormone receptors. Environ Health Perspect 113, 871-876. doi:10.1289/ehp.7793
    • (2005) Environ Health Perspect , vol.113 , pp. 871-876
    • Fritsche, E.1    Cline, J. E.2    Nguyen, N.-H.3
  • 19
    • 85018644461 scopus 로고    scopus 로고
    • OECD/EFSA workshop on developmental neurotoxicity (DNT): The use of non-animal test methods for regulatory purposes
    • Fritsche, E., Crofton, K. M., Hernandez, A. F. et al. (2017). OECD/EFSA workshop on developmental neurotoxicity (DNT): The use of non-animal test methods for regulatory purposes. ALTEX 34, 311-315. doi:10.14573/altex.1701171
    • (2017) ALTEX , vol.34 , pp. 311-315
    • Fritsche, E.1    Crofton, K. M.2    Hernandez, A. F.3
  • 20
    • 33845424655 scopus 로고    scopus 로고
    • Developmental neurotoxicity of industrial chemicals
    • Grandjean, P. and Landrigan, P. J. (2006). Developmental neurotoxicity of industrial chemicals. Lancet 368, 2167-2178. doi:10.1016/S0140-6736(06)69665-7
    • (2006) Lancet , vol.368 , pp. 2167-2178
    • Grandjean, P.1    Landrigan, P. J.2
  • 21
    • 84894029844 scopus 로고    scopus 로고
    • Neurobehavioural effects of developmental toxicity
    • Grandjean, P. and Landrigan, P. J. (2014). Neurobehavioural effects of developmental toxicity. Lancet Neurol 13, 330-338. doi:10.1016/S1474-4422(13)70278-3
    • (2014) Lancet Neurol , vol.13 , pp. 330-338
    • Grandjean, P.1    Landrigan, P. J.2
  • 22
    • 80052941505 scopus 로고    scopus 로고
    • Quantitative assessment of neurite outgrowth in PC12 cells
    • Harrill, J. A. and Mundy, W. R. (2011). Quantitative assessment of neurite outgrowth in PC12 cells. Methods Mol Biol 758, 331-348. doi:10.1007/978-1-61779-170-3_23
    • (2011) Methods Mol Biol , vol.758 , pp. 331-348
    • Harrill, J. A.1    Mundy, W. R.2
  • 23
    • 80855131654 scopus 로고    scopus 로고
    • Comparative sensitivity of human and rat neural cultures to chemical-induced inhibition of neurite outgrowth
    • Harrill, J. A., Freudenrich, T. M., Robinette, B. L. and Mundy, W. R. (2011). Comparative sensitivity of human and rat neural cultures to chemical-induced inhibition of neurite outgrowth. Toxicol Appl Pharmacol 256, 268-280. doi:10.1016/j. taap.2011.02.013
    • (2011) Toxicol Appl Pharmacol , vol.256 , pp. 268-280
    • Harrill, J. A.1    Freudenrich, T. M.2    Robinette, B. L.3    Mundy, W. R.4
  • 24
    • 84870357944 scopus 로고    scopus 로고
    • Use of high content image analyses to detect chemical-mediated effects on neurite sub-populations in primary rat cortical neurons
    • Harrill, J. A., Robinette, B. L., Freudenrich, T. and Mundy, W. R. (2013). Use of high content image analyses to detect chemical-mediated effects on neurite sub-populations in primary rat cortical neurons. Neurotoxicology 34, 61-73. doi:10.1016/j. neuro.2012.10.013
    • (2013) Neurotoxicology , vol.34 , pp. 61-73
    • Harrill, J. A.1    Robinette, B. L.2    Freudenrich, T.3    Mundy, W. R.4
  • 25
    • 84876323285 scopus 로고    scopus 로고
    • Food for thought … Integrated testing strategies for safety assessments
    • Hartung, T., Luechtefeld, T., Maertens, A. and Kleensang, A. (2013). Food for thought … Integrated testing strategies for safety assessments. ALTEX 30, 3-18. doi:10.14573/ altex.2013.1.003
    • (2013) ALTEX , vol.30 , pp. 3-18
    • Hartung, T.1    Luechtefeld, T.2    Maertens, A.3    Kleensang, A.4
  • 26
    • 84961258170 scopus 로고    scopus 로고
    • Stem cell-derived immature human dorsal root ganglia neurons to identify peripheral neurotoxicants
    • Hoelting, L., Klima, S., Karreman, C. et al. (2016). Stem cell-derived immature human dorsal root ganglia neurons to identify peripheral neurotoxicants. Stem Cells Transl Med 5, 476-487. doi:10.5966/sctm.2015-0108
    • (2016) Stem Cells Transl Med , vol.5 , pp. 476-487
    • Hoelting, L.1    Klima, S.2    Karreman, C.3
  • 27
    • 61349100071 scopus 로고    scopus 로고
    • Gene expression as a sensitive endpoint to evaluate cell differentiation and maturation of the developing central nervous system in primary cultures of rat cerebellar granule cells (CGCs) exposed to pesticides
    • Hogberg, H. T., Kinsner-Ovaskainen, A., Hartung, T. et al. (2009). Gene expression as a sensitive endpoint to evaluate cell differentiation and maturation of the developing central nervous system in primary cultures of rat cerebellar granule cells (CGCs) exposed to pesticides. Toxicol Appl Pharmacol 235, 268-286. doi:10.1016/j.taap.2008.12.014
    • (2009) Toxicol Appl Pharmacol , vol.235 , pp. 268-286
    • Hogberg, H. T.1    Kinsner-Ovaskainen, A.2    Hartung, T.3
  • 28
    • 75249106472 scopus 로고    scopus 로고
    • mRNA expression is a relevant tool to identify developmental neurotoxicants using an in vitro approach
    • Hogberg, H. T., Kinsner-Ovaskainen, A., Coecke, S. et al. (2010). mRNA expression is a relevant tool to identify developmental neurotoxicants using an in vitro approach. Toxicol Sci 113, 95-115. doi:10.1093/toxsci/kfp175
    • (2010) Toxicol Sci , vol.113 , pp. 95-115
    • Hogberg, H. T.1    Kinsner-Ovaskainen, A.2    Coecke, S.3
  • 29
    • 23744456959 scopus 로고    scopus 로고
    • Chlorpyrifos exerts opposing effects on axonal and dendritic growth in primary neuronal cultures
    • Howard, A. S., Bucelli, R., Jett, D. A. et al. (2005). Chlorpyrifos exerts opposing effects on axonal and dendritic growth in primary neuronal cultures. Toxicol Appl Pharmacol 207, 112-124. doi:10.1016/j.taap.2004.12.008
    • (2005) Toxicol Appl Pharmacol , vol.207 , pp. 112-124
    • Howard, A. S.1    Bucelli, R.2    Jett, D. A.3
  • 30
    • 79955724481 scopus 로고    scopus 로고
    • Integrated testing strategy (ITS) – Opportunities to better use existing data and guide future testing in toxicology
    • Jaworska, J. and Hoffmann, S. (2010). Integrated testing strategy (ITS) – Opportunities to better use existing data and guide future testing in toxicology. ALTEX 27, 231-242. doi:10.14573/altex.2010.4.231
    • (2010) ALTEX , vol.27 , pp. 231-242
    • Jaworska, J.1    Hoffmann, S.2
  • 31
    • 84902120685 scopus 로고    scopus 로고
    • In vitro and modelling approaches to risk assessment from the U.S. Environmental protection agency toxcast programme
    • Judson, R., Houck, K., Martin, M. et al. (2014). In vitro and modelling approaches to risk assessment from the U.S. Environmental protection agency toxcast programme. Basic Clin Pharmacol Toxicol 115, 69-76. doi:10.1111/bcpt.12239
    • (2014) Basic Clin Pharmacol Toxicol , vol.115 , pp. 69-76
    • Judson, R.1    Houck, K.2    Martin, M.3
  • 32
    • 84947781653 scopus 로고    scopus 로고
    • Integrated model of chemical perturbations of a biological pathway using 18 in vitro high-throughput screening assays for the estrogen receptor
    • Judson, R. S., Magpantay, F. M., Chickarmane, V. et al. (2015). Integrated model of chemical perturbations of a biological pathway using 18 in vitro high-throughput screening assays for the estrogen receptor. Toxicol Sci 148, 137-154. doi:10.1093/toxsci/kfv168
    • (2015) Toxicol Sci , vol.148 , pp. 137-154
    • Judson, R. S.1    Magpantay, F. M.2    Chickarmane, V.3
  • 33
    • 84992690576 scopus 로고    scopus 로고
    • Analysis of the effects of cell stress and cytotoxicity on in vitro assay activity across a diverse chemical and assay space
    • Judson, R., Houck, K., Martin, M. et al. (2016). Analysis of the effects of cell stress and cytotoxicity on in vitro assay activity across a diverse chemical and assay space. Toxicol Sci 152, 323-339. doi:10.1093/toxsci/kfw092
    • (2016) Toxicol Sci , vol.152 , pp. 323-339
    • Judson, R.1    Houck, K.2    Martin, M.3
  • 34
    • 79951679077 scopus 로고    scopus 로고
    • A core in vitro genotoxicity battery comprising the Ames test plus the in vitro micronucleus test is sufficient to detect rodent carcinogens and in vivo genotoxins
    • Kirkland, D., Reeve, L., Gatehouse, D. et al. (2011). A core in vitro genotoxicity battery comprising the Ames test plus the in vitro micronucleus test is sufficient to detect rodent carcinogens and in vivo genotoxins. Mutat Res 721, 27-73. doi:10.1016/j.mrgentox.2010.12.015
    • (2011) Mutat Res , vol.721 , pp. 27-73
    • Kirkland, D.1    Reeve, L.2    Gatehouse, D.3
  • 35
    • 84890566643 scopus 로고    scopus 로고
    • Evaluation of a human neurite growth assay as specific screen for developmental neurotoxicants
    • Krug, A. K., Balmer, N. V., Matt, F. et al. (2013). Evaluation of a human neurite growth assay as specific screen for developmental neurotoxicants. Arch Toxicol 87, 2215-2231. doi:10.1007/s00204-013-1072-y
    • (2013) Arch Toxicol , vol.87 , pp. 2215-2231
    • Krug, A. K.1    Balmer, N. V.2    Matt, F.3
  • 37
    • 59849124092 scopus 로고    scopus 로고
    • The dawning of a new age of toxicology
    • Leist, M., Hartung, T. and Nicotera, P. (2008a). The dawning of a new age of toxicology. ALTEX 25, 103-114. doi:10.14573/ altex.2008.2.103
    • (2008) ALTEX , vol.25 , pp. 103-114
    • Leist, M.1    Hartung, T.2    Nicotera, P.3
  • 38
    • 48949091622 scopus 로고    scopus 로고
    • Food for thought ... On the real success of 3R approaches
    • Leist, M., Kadereit, S. and Schildknecht, S. (2008b). Food for thought ... On the real success of 3R approaches. ALTEX 25, 17-32. doi:10.14573/altex.2008.1.17
    • (2008) ALTEX , vol.25 , pp. 17-32
    • Leist, M.1    Kadereit, S.2    Schildknecht, S.3
  • 39
    • 79955432219 scopus 로고    scopus 로고
    • Food for thought ... Considerations and guidelines for basic test method descriptions in toxicology
    • Leist, M., Efremova, L. and Karreman, C. (2010). Food for thought ... Considerations and guidelines for basic test method descriptions in toxicology. ALTEX 27, 309-317. doi:10.14573/altex.2010.4.309
    • (2010) ALTEX , vol.27 , pp. 309-317
    • Leist, M.1    Efremova, L.2    Karreman, C.3
  • 40
    • 84864593532 scopus 로고    scopus 로고
    • Validation and quality control of replacement alternatives – Current status and future challenges
    • Leist, M., Hasiwa, N., Daneshian, M. and Hartung, T. (2012). Validation and quality control of replacement alternatives – Current status and future challenges. Toxicol Res 1, 8-22. doi:10.1039/c2tx20011b
    • (2012) Toxicol Res , vol.1 , pp. 8-22
    • Leist, M.1    Hasiwa, N.2    Daneshian, M.3    Hartung, T.4
  • 41
    • 84904901979 scopus 로고    scopus 로고
    • Consensus report on the future of animal-free systemic toxicity testing
    • Leist, M., Hasiwa, N., Rovida, C. et al. (2014). Consensus report on the future of animal-free systemic toxicity testing. ALTEX 31, 341-356. doi:10.14573/altex.1406091
    • (2014) ALTEX , vol.31 , pp. 341-356
    • Leist, M.1    Hasiwa, N.2    Rovida, C.3
  • 42
    • 85035790758 scopus 로고    scopus 로고
    • The borderline range of toxicological methods: Quantification and implications for evaluating precision
    • Leontaridou, M., Urbisch, D., Kolle, S. N. et al. (2017). The borderline range of toxicological methods: Quantification and implications for evaluating precision. ALTEX 34, 525-538. doi:10.14573/altex.1606271
    • (2017) ALTEX , vol.34 , pp. 525-538
    • Leontaridou, M.1    Urbisch, D.2    Kolle, S. N.3
  • 43
    • 0037064078 scopus 로고    scopus 로고
    • Effect of mutant alpha-synuclein on dopamine homeostasis in a new human mesencephalic cell line
    • Lotharius, J., Barg, S., Wiekop, P. et al. (2002). Effect of mutant alpha-synuclein on dopamine homeostasis in a new human mesencephalic cell line. J Biol Chem 277, 38884-38894. doi:10.1074/jbc.M205518200
    • (2002) J Biol Chem , vol.277 , pp. 38884-38894
    • Lotharius, J.1    Barg, S.2    Wiekop, P.3
  • 44
    • 21844442345 scopus 로고    scopus 로고
    • Progressive degeneration of human mesencephalic neuron-derived cells triggered by dopamine-dependent oxidative stress is dependent on the mixed-lineage kinase pathway
    • Lotharius, J., Falsig, J., van Beek, J. et al. (2005). Progressive degeneration of human mesencephalic neuron-derived cells triggered by dopamine-dependent oxidative stress is dependent on the mixed-lineage kinase pathway. J Neurosci 25, 6329-6342. doi:10.1523/JNEUROSCI.1746-05.2005
    • (2005) J Neurosci , vol.25 , pp. 6329-6342
    • Lotharius, J.1    Falsig, J.2    van Beek, J.3
  • 45
    • 60749100386 scopus 로고    scopus 로고
    • A retrospective performance assessment of the developmental neurotoxicity study in support of OECD test guideline 426
    • Makris, S. L., Raffaele, K., Allen, S. et al. (2009). A retrospective performance assessment of the developmental neurotoxicity study in support of OECD test guideline 426. Environ Health Perspect 117, 17-25. doi:10.1289/ehp.11447
    • (2009) Environ Health Perspect , vol.117 , pp. 17-25
    • Makris, S. L.1    Raffaele, K.2    Allen, S.3
  • 46
    • 84979255712 scopus 로고    scopus 로고
    • Biology-inspired microphysiological system approaches to solve the prediction dilemma of substance testing
    • Marx, U., Andersson, T. B., Bahinski, A. et al. (2016). Biology-inspired microphysiological system approaches to solve the prediction dilemma of substance testing. ALTEX 33, 272-321. doi:10.14573/altex.1603161
    • (2016) ALTEX , vol.33 , pp. 272-321
    • Marx, U.1    Andersson, T. B.2    Bahinski, A.3
  • 47
    • 37249023309 scopus 로고    scopus 로고
    • New and original pKa prediction method using grid molecular interaction fields
    • Milletti, F., Storchi, L., Sforna, G. and Cruciani, G. (2007). New and original pKa prediction method using grid molecular interaction fields. J Chem Inf Model 47, 2172-2181. doi:10.1021/ci700018y
    • (2007) J Chem Inf Model , vol.47 , pp. 2172-2181
    • Milletti, F.1    Storchi, L.2    Sforna, G.3    Cruciani, G.4
  • 48
    • 84944718074 scopus 로고    scopus 로고
    • Expanding the test set: Chemicals with potential to disrupt mammalian brain development
    • Mundy, W. R., Padilla, S., Breier, J. M. et al. (2015). Expanding the test set: Chemicals with potential to disrupt mammalian brain development. Neurotoxicol Teratol 52, 25-35. doi:10.1016/j.ntt.2015.10.001
    • (2015) Neurotoxicol Teratol , vol.52 , pp. 25-35
    • Mundy, W. R.1    Padilla, S.2    Breier, J. M.3
  • 49
    • 85018724518 scopus 로고    scopus 로고
    • Combination of multiple neural crest migration assays to identify environmental toxicants from a proof-of-concept chemical library
    • Nyffeler, J., Dolde, X., Krebs, A. et al. (2017a). Combination of multiple neural crest migration assays to identify environmental toxicants from a proof-of-concept chemical library. Arch Toxicol 91, 3613–3632. doi:10.1007/s00204-017-1977-y
    • (2017) Arch Toxicol , vol.91 , pp. 3613-3632
    • Nyffeler, J.1    Dolde, X.2    Krebs, A.3
  • 50
    • 85012008949 scopus 로고    scopus 로고
    • Design of a high-throughput human neural crest cell migration assay to indicate potential developmental toxicants
    • Nyffeler, J., Karreman, C., Leisner, H. et al. (2017b). Design of a high-throughput human neural crest cell migration assay to indicate potential developmental toxicants. ALTEX 34, 75-94. doi:10.14573/altex.1605031
    • (2017) ALTEX , vol.34 , pp. 75-94
    • Nyffeler, J.1    Karreman, C.2    Leisner, H.3
  • 52
    • 85026243203 scopus 로고    scopus 로고
    • A human brain microphysiological system derived from induced pluripotent stem cells to study neurological diseases and toxicity
    • Pamies, D., Barreras, P., Block, K. et al. (2016). A human brain microphysiological system derived from induced pluripotent stem cells to study neurological diseases and toxicity. ALTEX 34, 362-376. doi:10.14573/altex.1609122
    • (2016) ALTEX , vol.34 , pp. 362-376
    • Pamies, D.1    Barreras, P.2    Block, K.3
  • 53
    • 84877604097 scopus 로고    scopus 로고
    • Uncertainty of testing methods – What do we (want to) know?
    • Paparella, M., Daneshian, M., Hornek-Gausterer, R. et al. (2013). Uncertainty of testing methods – What do we (want to) know? ALTEX 30, 131-144. doi:10.14573/altex.2013.2.131
    • (2013) ALTEX , vol.30 , pp. 131-144
    • Paparella, M.1    Daneshian, M.2    Hornek-Gausterer, R.3
  • 54
    • 85018622397 scopus 로고    scopus 로고
    • Uncertainties of testing methods: What do we (want to) know about carcinogenicity?
    • Paparella, M., Colacci, A. and Jacobs, M. N. (2017). Uncertainties of testing methods: What do we (want to) know about carcinogenicity? ALTEX 34, 235-252. doi:10.14573/ altex.1608281
    • (2017) ALTEX , vol.34 , pp. 235-252
    • Paparella, M.1    Colacci, A.2    Jacobs, M. N.3
  • 55
    • 0034710718 scopus 로고    scopus 로고
    • Grid-independent descriptors (GRIND): A novel class of alignment-independent three-dimensional molecular descriptors
    • Pastor, M., Cruciani, G., McLay, I. et al. (2000). Grid-independent descriptors (GRIND): A novel class of alignment-independent three-dimensional molecular descriptors. J Med Chem 43, 3233-3243. doi:10.1021/jm000941m
    • (2000) J Med Chem , vol.43 , pp. 3233-3243
    • Pastor, M.1    Cruciani, G.2    McLay, I.3
  • 56
    • 84939857473 scopus 로고    scopus 로고
    • Comparative neurotoxicity screening in human iPSC-derived neural stem cells, neurons and astrocytes
    • Pei, Y., Peng, J., Behl, M. et al. (2016). Comparative neurotoxicity screening in human iPSC-derived neural stem cells, neurons and astrocytes. Brain Res 1638, 57-73. doi:10.1016/j. brainres.2015.07.048
    • (2016) Brain Res , vol.1638 , pp. 57-73
    • Pei, Y.1    Peng, J.2    Behl, M.3
  • 57
    • 85013231143 scopus 로고    scopus 로고
    • The isolated chicken eye test to replace the Draize test in rabbits
    • Prinsen, M. K., Hendriksen, C. F., Krul, C. A. and Woutersen, R. A. (2017). The isolated chicken eye test to replace the Draize test in rabbits. Regul Toxicol Pharmacol 85, 132-149. doi:10.1016/j.yrtph.2017.01.009
    • (2017) Regul Toxicol Pharmacol , vol.85 , pp. 132-149
    • Prinsen, M. K.1    Hendriksen, C. F.2    Krul, C. A.3    Woutersen, R. A.4
  • 58
    • 84959358615 scopus 로고    scopus 로고
    • Neurite outgrowth in human induced pluripotent stem cell-derived neurons as a high-throughput screen for developmental neurotoxicity or neurotoxicity
    • Ryan, K. R., Sirenko, O., Parham, F. et al. (2016). Neurite outgrowth in human induced pluripotent stem cell-derived neurons as a high-throughput screen for developmental neurotoxicity or neurotoxicity. Neurotoxicology 53, 271-281. doi:10.1016/j.neuro.2016.02.003
    • (2016) Neurotoxicology , vol.53 , pp. 271-281
    • Ryan, K. R.1    Sirenko, O.2    Parham, F.3
  • 59
    • 0028466540 scopus 로고
    • Comparison of automatic three-dimensional model builders using 639 x-ray structures
    • Sadowski, J., Gasteiger, J. and Klebe, G. (1994). Comparison of automatic three-dimensional model builders using 639 x-ray structures. J Chem Inf Comput Sci 34, 1000-1008. doi:10.1021/ci00020a039
    • (1994) J Chem Inf Comput Sci , vol.34 , pp. 1000-1008
    • Sadowski, J.1    Gasteiger, J.2    Klebe, G.3
  • 60
    • 70350031056 scopus 로고    scopus 로고
    • Requirement of a dopaminergic neuronal phenotype for toxicity of low concentrations of 1-methyl-4-phenylpyridinium to human cells
    • Schildknecht, S., Poltl, D., Nagel, D. M. et al. (2009). Requirement of a dopaminergic neuronal phenotype for toxicity of low concentrations of 1-methyl-4-phenylpyridinium to human cells. Toxicol Appl Pharmacol 241, 23-35. doi:10.1016/j. taap.2009.07.027
    • (2009) Toxicol Appl Pharmacol , vol.241 , pp. 23-35
    • Schildknecht, S.1    Poltl, D.2    Nagel, D. M.3
  • 61
    • 84893041165 scopus 로고    scopus 로고
    • Generation of genetically-modified human differentiated cells for toxicological tests and the study of neurodegenerative diseases
    • Schildknecht, S., Karreman, C., Poltl, D. et al. (2013). Generation of genetically-modified human differentiated cells for toxicological tests and the study of neurodegenerative diseases. ALTEX 30, 427-444. doi:10.14573/altex.2013.4.427
    • (2013) ALTEX , vol.30 , pp. 427-444
    • Schildknecht, S.1    Karreman, C.2    Poltl, D.3
  • 62
    • 85018794797 scopus 로고    scopus 로고
    • Tipping points and endogenous determinants of nigrostriatal degeneration by MPTP
    • Schildknecht, S., Di Monte, D. A., Pape, R. et al. (2017). Tipping points and endogenous determinants of nigrostriatal degeneration by MPTP. Trends Pharmacol Sci 38, 541-555. doi:10.1016/j.tips.2017.03.010
    • (2017) Trends Pharmacol Sci , vol.38 , pp. 541-555
    • Schildknecht, S.1    Di Monte, D. A.2    Pape, R.3
  • 63
    • 84983002040 scopus 로고    scopus 로고
    • In vitro acute and developmental neurotoxicity screening: An overview of cellular platforms and high-throughput technical possibilities
    • Schmidt, B. Z., Lehmann, M., Gutbier, S. et al. (2017). In vitro acute and developmental neurotoxicity screening: An overview of cellular platforms and high-throughput technical possibilities. Arch Toxicol 91, 1-33. doi:10.1007/s00204-016-1805-9
    • (2017) Arch Toxicol , vol.91 , pp. 1-33
    • Schmidt, B. Z.1    Lehmann, M.2    Gutbier, S.3
  • 64
    • 84990890584 scopus 로고    scopus 로고
    • Omnisphero: A high-content image analysis (HCA) approach for phenotypic developmental neurotoxicity (DNT) screenings of organoid neurosphere cultures in vitro
    • Schmuck, M. R., Temme, T., Dach, K. et al. (2017). Omnisphero: A high-content image analysis (HCA) approach for phenotypic developmental neurotoxicity (DNT) screenings of organoid neurosphere cultures in vitro. Arch Toxicol 91, 2017-2028. doi:10.1007/s00204-016-1852-2
    • (2017) Arch Toxicol , vol.91 , pp. 2017-2028
    • Schmuck, M. R.1    Temme, T.2    Dach, K.3
  • 65
    • 81055127451 scopus 로고    scopus 로고
    • Rapid, complete and large-scale generation of post-mitotic neurons from the human LUHMES cell line
    • Scholz, D., Poltl, D., Genewsky, A. et al. (2011). Rapid, complete and large-scale generation of post-mitotic neurons from the human LUHMES cell line. J Neurochem 119, 957-971. doi:10.1111/j.1471-4159.2011.07255.x
    • (2011) J Neurochem , vol.119 , pp. 957-971
    • Scholz, D.1    Poltl, D.2    Genewsky, A.3
  • 66
    • 84969242925 scopus 로고    scopus 로고
    • Definition of transcriptome-based indices for quantitative characterization of chemically disturbed stem cell development: Introduction of the STOP-Toxukn and STOP-Toxukk tests
    • Shinde, V., Hoelting, L., Srinivasan, S. P. et al. (2017). Definition of transcriptome-based indices for quantitative characterization of chemically disturbed stem cell development: Introduction of the STOP-Toxukn and STOP-Toxukk tests. Arch Toxicol 91, 839-864. doi:10.1007/s00204-016-1741-8
    • (2017) Arch Toxicol , vol.91 , pp. 839-864
    • Shinde, V.1    Hoelting, L.2    Srinivasan, S. P.3
  • 67
    • 85015061049 scopus 로고    scopus 로고
    • In vitro cardiotoxicity assessment of environmental chemicals using an organotypic human induced pluripotent stem cell-derived model
    • Sirenko, O., Grimm, F. A., Ryan, K. R. et al. (2017). In vitro cardiotoxicity assessment of environmental chemicals using an organotypic human induced pluripotent stem cell-derived model. Toxicol Appl Pharmacol 322, 60-74. doi:10.1016/j. taap.2017.02.020
    • (2017) Toxicol Appl Pharmacol , vol.322 , pp. 60-74
    • Sirenko, O.1    Grimm, F. A.2    Ryan, K. R.3
  • 69
    • 84949509769 scopus 로고    scopus 로고
    • A LUHMES 3D dopaminergic neuronal model for neurotoxicity testing allowing long-term exposure and cellular resilience analysis
    • Smirnova, L., Harris, G., Delp, J. et al. (2016). A LUHMES 3D dopaminergic neuronal model for neurotoxicity testing allowing long-term exposure and cellular resilience analysis. Arch Toxicol 90, 2725-2743. doi:10.1007/s00204-015-1637-z
    • (2016) Arch Toxicol , vol.90 , pp. 2725-2743
    • Smirnova, L.1    Harris, G.2    Delp, J.3
  • 70
    • 0016593776 scopus 로고
    • Nervous system degeneration produced by acrylamide monomer
    • Spencer, P. S. and Schaumburg, H. H. (1975). Nervous system degeneration produced by acrylamide monomer. Environ Health Perspect 11, 129-133. doi:10.1289/ehp.7511129
    • (1975) Environ Health Perspect , vol.11 , pp. 129-133
    • Spencer, P. S.1    Schaumburg, H. H.2
  • 71
    • 79955379993 scopus 로고    scopus 로고
    • Assessment of chemical-induced impairment of human neurite outgrowth by multiparametric live cell imaging in high-density cultures
    • Stiegler, N. V., Krug, A. K., Matt, F. and Leist, M. (2011). Assessment of chemical-induced impairment of human neurite outgrowth by multiparametric live cell imaging in high-density cultures. Toxicol Sci 121, 73-87. doi:10.1093/toxsci/kfr034
    • (2011) Toxicol Sci , vol.121 , pp. 73-87
    • Stiegler, N. V.1    Krug, A. K.2    Matt, F.3    Leist, M.4
  • 72
    • 0032491416 scopus 로고    scopus 로고
    • Embryonic stem cell lines derived from human blastocysts
    • Thomson, J. A., Itskovitz-Eldor, J., Shapiro, S. S. et al. (1998). Embryonic stem cell lines derived from human blastocysts. Science 282, 1145-1147. doi:10.1126/science.282.5391. 1145
    • (1998) Science , vol.282 , pp. 1145-1147
    • Thomson, J. A.1    Itskovitz-Eldor, J.2    Shapiro, S. S.3
  • 73
    • 84879599189 scopus 로고    scopus 로고
    • Improving the human hazard characterization of chemicals: A Tox21 update
    • Tice, R. R., Austin, C. P., Kavlock, R. J. and Bucher, J. R. (2013). Improving the human hazard characterization of chemicals: A Tox21 update. Environ Health Perspect 121, 756-765. doi:10.1289/ehp.1205784
    • (2013) Environ Health Perspect , vol.121 , pp. 756-765
    • Tice, R. R.1    Austin, C. P.2    Kavlock, R. J.3    Bucher, J. R.4
  • 74
    • 84864511648 scopus 로고    scopus 로고
    • Translating neurobehavioural endpoints of developmental neurotoxicity tests into in vitro assays and readouts
    • van Thriel, C., Westerink, R. H., Beste, C. et al. (2012). Translating neurobehavioural endpoints of developmental neurotoxicity tests into in vitro assays and readouts. Neurotoxicology 33, 911-924. doi:10.1016/j.neuro.2011.10.002
    • (2012) Neurotoxicology , vol.33 , pp. 911-924
    • van Thriel, C.1    Westerink, R. H.2    Beste, C.3
  • 75
    • 38549151817 scopus 로고    scopus 로고
    • Drugbank: A knowledgebase for drugs, drug actions and drug targets
    • Wishart, D. S., Knox, C., Guo, A. C. et al. (2008). Drugbank: A knowledgebase for drugs, drug actions and drug targets. Nucleic Acids Res 36, D901-D906. doi:10.1093/nar/gkm958
    • (2008) Nucleic Acids Res , vol.36 , pp. D901-D906
    • Wishart, D. S.1    Knox, C.2    Guo, A. C.3
  • 76
    • 84894262438 scopus 로고    scopus 로고
    • PCB 136 atropselectively alters morphometric and functional parameters of neuronal connectivity in cultured rat hippocampal neurons via ryanodine receptor-dependent mechanisms
    • Yang, D., Kania-Korwel, I., Ghogha, A. et al. (2014). PCB 136 atropselectively alters morphometric and functional parameters of neuronal connectivity in cultured rat hippocampal neurons via ryanodine receptor-dependent mechanisms. Toxicol Sci 138, 379-392. doi:10.1093/toxsci/kft334
    • (2014) Toxicol Sci , vol.138 , pp. 379-392
    • Yang, D.1    Kania-Korwel, I.2    Ghogha, A.3
  • 77
    • 79951678616 scopus 로고    scopus 로고
    • Coordinated waves of gene expression during neuronal differentiation of embryonic stem cells as basis for novel approaches to developmental neurotoxicity testing
    • Zimmer, B., Kuegler, P. B., Baudis, B. et al. (2011a). Coordinated waves of gene expression during neuronal differentiation of embryonic stem cells as basis for novel approaches to developmental neurotoxicity testing. Cell Death Differ 18, 383-395. doi:10.1038/cdd.2010.109
    • (2011) Cell Death Differ , vol.18 , pp. 383-395
    • Zimmer, B.1    Kuegler, P. B.2    Baudis, B.3
  • 78
    • 79957868414 scopus 로고    scopus 로고
    • Sensitivity of dopaminergic neuron differentiation from stem cells to chronic low-dose methylmercury exposure
    • Zimmer, B., Schildknecht, S., Kuegler, P. B. et al. (2011b). Sensitivity of dopaminergic neuron differentiation from stem cells to chronic low-dose methylmercury exposure. Toxicol Sci 121, 357-367. doi:10.1093/toxsci/kfr054
    • (2011) Toxicol Sci , vol.121 , pp. 357-367
    • Zimmer, B.1    Schildknecht, S.2    Kuegler, P. B.3
  • 79
    • 84864758119 scopus 로고    scopus 로고
    • Evaluation of developmental toxicants and signaling pathways in a functional test based on the migration of human neural crest cells
    • Zimmer, B., Lee, G., Balmer, N. V. et al. (2012). Evaluation of developmental toxicants and signaling pathways in a functional test based on the migration of human neural crest cells. Environ Health Perspect 120, 1116-1122. doi:10.1289/ ehp.1104489
    • (2012) Environ Health Perspect , vol.120 , pp. 1116-1122
    • Zimmer, B.1    Lee, G.2    Balmer, N. V.3
  • 80
    • 84901307535 scopus 로고    scopus 로고
    • Profiling of drugs and environmental chemicals for functional impairment of neural crest migration in a novel stem cell-based test battery
    • Zimmer, B., Pallocca, G., Dreser, N. et al. (2014). Profiling of drugs and environmental chemicals for functional impairment of neural crest migration in a novel stem cell-based test battery. Arch Toxicol 88, 1109-1126. doi:10.1007/s00204-014-1231-9
    • (2014) Arch Toxicol , vol.88 , pp. 1109-1126
    • Zimmer, B.1    Pallocca, G.2    Dreser, N.3


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.