메뉴 건너뛰기




Volumn 35, Issue 3, 2018, Pages 306-352

Recommendation on Test Readiness Criteria for New Approach Methods in Toxicology: Exemplified for Developmental Neurotoxicity

(21)  Bal Price, Anna a   Hogberg, Helena T b   Crofton, Kevin M c   Daneshian, Mardas d   FitzGerald, Rex E e   Fritsche, Ellen f   Heinonen, Tuula g   Bennekou, Susanne Hougaard h   Klima, Stefanie d   Piersma, Aldert H i   Sachana, Magdalini j   Shafer, Timothy J c   Terron, Andrea k   Monnet Tschudi, Florianne e,l   Viviani, Barbara m   Waldmann, Tanja d   Westerink, Remco H S i   Wilks, Martin F e   Witters, Hilda n   Zurich, Marie Gabrielle e,l   more..


Author keywords

[No Author keywords available]

Indexed keywords

ANIMAL; ANIMAL TESTING ALTERNATIVE; EDUCATION; HUMAN; PRACTICE GUIDELINE; PROCEDURES; RISK ASSESSMENT; TOXICITY AND INTOXICATION; TOXICITY TESTING; TRENDS;

EID: 85051034983     PISSN: 1868596X     EISSN: 18688551     Source Type: Journal    
DOI: 10.14573/altex.1712081     Document Type: Article
Times cited : (138)

References (366)
  • 1
    • 0025231659 scopus 로고
    • Neonatal hyperthyroidism in the rat produces an increase in the activity of microperoxisomal marker enzymes coincident with biochemical signs of accelerated myelination
    • Adamo, A. M., Aloise, P. A., Soto, E. F. and Pasquini, J. M. (1990). Neonatal hyperthyroidism in the rat produces an increase in the activity of microperoxisomal marker enzymes coincident with biochemical signs of accelerated myelination. J Neurosci Res 25, 353-359. doi:10.1002/jnr.490250312
    • (1990) J Neurosci Res , vol.25 , pp. 353-359
    • Adamo, A. M.1    Aloise, P. A.2    Soto, E. F.3    Pasquini, J. M.4
  • 2
    • 84859019118 scopus 로고    scopus 로고
    • All-trans-retinoid acid induces the differentiation of encapsulated mouse embryonic stem cells into GABAergic neurons
    • Addae, C., Yi, X., Gernapudi, R. et al. (2012). All-trans-retinoid acid induces the differentiation of encapsulated mouse embryonic stem cells into GABAergic neurons. Differentiation 83, 233-241. doi:10.1016/j.diff.2012.03.001
    • (2012) Differentiation , vol.83 , pp. 233-241
    • Addae, C.1    Yi, X.2    Gernapudi, R.3
  • 3
    • 84930091540 scopus 로고    scopus 로고
    • st century (TT21C): Making safety decisions using toxicity pathways, and progress in a prototype risk assessment
    • st century (TT21C): Making safety decisions using toxicity pathways, and progress in a prototype risk assessment. Toxicology 332, 102-111. doi:10.1016/j. tox.2014.02.007
    • (2015) Toxicology , vol.332 , pp. 102-111
    • Adeleye, Y.1    Andersen, M.2    Clewell, R.3
  • 4
    • 84910682457 scopus 로고    scopus 로고
    • State-of-the-art of 3D cultures (organs-on-a-chip) in safety testing and pathophysiology
    • Alépée, N., Bahinski, A., Daneshian, M. et al. (2014). State-of-the-art of 3D cultures (organs-on-a-chip) in safety testing and pathophysiology. ALTEX 31, 441-477. doi:10.14573/ altex1406111
    • (2014) ALTEX , vol.31 , pp. 441-477
    • Alépée, N.1    Bahinski, A.2    Daneshian, M.3
  • 5
    • 84856224090 scopus 로고    scopus 로고
    • Behavioral profiling of zebrafish embryos exposed to a panel of 60 water-soluble compounds
    • Ali, S., Champagne, D. L. and Richardson, M. K. (2012). Behavioral profiling of zebrafish embryos exposed to a panel of 60 water-soluble compounds. Behav Brain Res 228, 272-283. doi:10.1016/j.bbr.2011.11.020
    • (2012) Behav Brain Res , vol.228 , pp. 272-283
    • Ali, S.1    Champagne, D. L.2    Richardson, M. K.3
  • 6
    • 77951118386 scopus 로고    scopus 로고
    • Adverse outcome pathways: A conceptual framework to support ecotoxicology research and risk assessment
    • Ankley, G. T., Bennett, R. S., Erickson, R. J. et al. (2010). Adverse outcome pathways: A conceptual framework to support ecotoxicology research and risk assessment. Environ Toxicol Chem 29, 730-741. doi:10.1002/etc.34
    • (2010) Environ Toxicol Chem , vol.29 , pp. 730-741
    • Ankley, G. T.1    Bennett, R. S.2    Erickson, R. J.3
  • 7
    • 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
  • 8
    • 84876694073 scopus 로고    scopus 로고
    • Ketamine enhances human neural stem cell proliferation and induces neuronal apoptosis via reactive oxygen species-mediated mitochondrial pathway
    • Bai, X., Yan, Y., Canfield, S. et al. (2013). Ketamine enhances human neural stem cell proliferation and induces neuronal apoptosis via reactive oxygen species-mediated mitochondrial pathway. Anesth Analg 116, 869-880. doi:10.1213/ ANE.0b013e3182860fc9
    • (2013) Anesth Analg , vol.116 , pp. 869-880
    • Bai, X.1    Yan, Y.2    Canfield, S.3
  • 9
    • 74249102208 scopus 로고    scopus 로고
    • Assessing the effects of the neonicotinoid insecticide imidacloprid in the cholinergic synapses of the stellate cells of the mouse cochlear nucleus using whole-cell patch-clamp recording
    • Bal, R., Erdogan, S., Theophilidis, G. et al. (2010). Assessing the effects of the neonicotinoid insecticide imidacloprid in the cholinergic synapses of the stellate cells of the mouse cochlear nucleus using whole-cell patch-clamp recording. Neurotoxicology 31, 113-120. doi:10.1016/j.neuro.2009.10.004
    • (2010) Neurotoxicology , vol.31 , pp. 113-120
    • Bal, R.1    Erdogan, S.2    Theophilidis, G.3
  • 10
    • 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., 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.2    Zimmer, B.3
  • 11
    • 84902118292 scopus 로고    scopus 로고
    • Epigenetics and transcriptomics to detect adverse drug effects in model systems of human development
    • Balmer, N. V. and Leist, M. (2014). Epigenetics and transcriptomics to detect adverse drug effects in model systems of human development. Basic Clin Pharmacol Toxicol 115, 59-68. doi:10.1111/bcpt.12203
    • (2014) Basic Clin Pharmacol Toxicol , vol.115 , pp. 59-68
    • Balmer, N. V.1    Leist, M.2
  • 12
    • 84903439132 scopus 로고    scopus 로고
    • From transient transcriptome responses to disturbed neurodevelopment: Role of histone acetylation and methylation as epigenetic switch between reversible and irreversible drug effects
    • Balmer, N. V., Klima, S., Rempel, E. et al. (2014). From transient transcriptome responses to disturbed neurodevelopment: Role of histone acetylation and methylation as epigenetic switch between reversible and irreversible drug effects. Arch Toxicol 88, 1451-1468. doi:10.1007/s00204-014-1279-6
    • (2014) Arch Toxicol , vol.88 , pp. 1451-1468
    • Balmer, N. V.1    Klima, S.2    Rempel, E.3
  • 13
    • 77956226283 scopus 로고    scopus 로고
    • In vitro developmental neurotoxicity (DNT) testing: Relevant models and endpoints
    • Bal-Price, A. K., Hogberg, H. T. and Buzanska, L. (2010). In vitro developmental neurotoxicity (DNT) testing: Relevant models and endpoints. Neurotoxicology 31, 545-554. doi:10.1016/j. neuro.2009.11.006
    • (2010) Neurotoxicology , vol.31 , pp. 545-554
    • Bal-Price, A. K.1    Hogberg, H. T.2    Buzanska, L.3
  • 14
    • 84860801327 scopus 로고    scopus 로고
    • Advancing the science of developmental neurotoxicity (DNT): Testing for better safety evaluation
    • Bal-Price, A. K., Coecke, S., Costa, L. et al. (2012). Advancing the science of developmental neurotoxicity (DNT): Testing for better safety evaluation. ALTEX 29, 202-215. doi:10.14573/ altex.2012.2.202
    • (2012) ALTEX , vol.29 , pp. 202-215
    • Bal-Price, A. K.1    Coecke, S.2    Costa, L.3
  • 15
    • 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. (2015a). 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
  • 16
    • 84921474981 scopus 로고    scopus 로고
    • Putative adverse outcome pathways relevant to neurotoxicity
    • Bal-Price, A., Crofton, K. M., Sachana, M. et al. (2015b). Putative adverse outcome pathways relevant to neurotoxicity. Crit Rev Toxicol 45, 83-91. doi:10.3109/10408444.2014.981331
    • (2015) Crit Rev Toxicol , vol.45 , pp. 83-91
    • Bal-Price, A.1    Crofton, K. M.2    Sachana, M.3
  • 17
    • 85020083235 scopus 로고    scopus 로고
    • Adverse outcome pathways: Application to enhance mechanistic understanding of neurotoxicity
    • Bal-Price, A. and Meek, M. E. B. (2017). Adverse outcome pathways: Application to enhance mechanistic understanding of neurotoxicity. Pharmacol Ther 179, 84-95. doi:10.1016/j. pharmthera.2017.05.006
    • (2017) Pharmacol Ther , vol.179 , pp. 84-95
    • Bal-Price, A.1    Meek, M. E. B.2
  • 18
    • 84973862422 scopus 로고    scopus 로고
    • Developing and applying the adverse outcome pathway concept for understanding and predicting neurotoxicity
    • Bal-Price, A., Lein, P. J., Keil, K. P. et al. (2017). Developing and applying the adverse outcome pathway concept for understanding and predicting neurotoxicity. Neurotoxicology 59, 240-255. doi:10.1016/j.neuro.2016.05.010
    • (2017) Neurotoxicology , vol.59 , pp. 240-255
    • Bal-Price, A.1    Lein, P. J.2    Keil, K. P.3
  • 19
    • 85011636463 scopus 로고    scopus 로고
    • Epigallocatechin gallate (EGCG) inhibits adhesion and migration of neural progenitor cells in vitro
    • Barenys, M., Gassmann, K., Baksmeier, C. et al. (2017). Epigallocatechin gallate (EGCG) inhibits adhesion and migration of neural progenitor cells in vitro. Arch Toxicol 91, 827-837. doi:10.1007/s00204-016-1709-8
    • (2017) Arch Toxicol , vol.91 , pp. 827-837
    • Barenys, M.1    Gassmann, K.2    Baksmeier, C.3
  • 20
    • 84902479918 scopus 로고    scopus 로고
    • Comparative human and rat “neurosphere assay” for developmental neurotoxicity testing
    • 12.21
    • Baumann, J., Barenys, M., Gassmann, K. and Fritsche, E. (2014). Comparative human and rat “neurosphere assay” for developmental neurotoxicity testing. Curr Protoc Toxicol 59, 12.21.1-24. doi:10.1002/0471140856.tx1221s59
    • (2014) Curr Protoc Toxicol , vol.59 , pp. 1-24
    • Baumann, J.1    Barenys, M.2    Gassmann, K.3    Fritsche, E.4
  • 21
    • 85144885421 scopus 로고    scopus 로고
    • Application of the neurosphere assay for DNT hazard assessment: Challenges and limitations
    • Humana Press
    • Baumann, J., Dach, K., Barenys, M. et al. (2015). Application of the neurosphere assay for DNT hazard assessment: Challenges and limitations. In Methods in Pharmacology and Toxicology. Humana Press. doi:10.1007/7653_2015_49
    • (2015) Methods in Pharmacology and Toxicology
    • Baumann, J.1    Dach, K.2    Barenys, M.3
  • 22
    • 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
  • 24
    • 84947764533 scopus 로고    scopus 로고
    • Rapid generation of sub-type, region specific neurons and neural networks from human pluripotent stem cell-derived neurospheres
    • Begum, A. N., Guoynes, C., Cho, J. et al. (2015). Rapid generation of sub-type, region specific neurons and neural networks from human pluripotent stem cell-derived neurospheres. Stem Cell Res 15, 731-741. doi:10.1016/j.scr.2015.10.014
    • (2015) Stem Cell Res , vol.15 , pp. 731-741
    • Begum, A. N.1    Guoynes, C.2    Cho, J.3
  • 25
    • 84951192820 scopus 로고    scopus 로고
    • Use of alternative assays to identify and prioritize organophosphorus flame retardants for potential developmental and neurotoxicity
    • Behl, M., Hsieh, J. H., Shafer, T. J. et al. (2015). Use of alternative assays to identify and prioritize organophosphorus flame retardants for potential developmental and neurotoxicity. Neurotoxicol Teratol 52, 181-193. doi:10.1016/j.ntt.2015.09. 003
    • (2015) Neurotoxicol Teratol , vol.52 , pp. 181-193
    • Behl, M.1    Hsieh, J. H.2    Shafer, T. J.3
  • 26
    • 85040628728 scopus 로고    scopus 로고
    • Editor’s highlight: Comparative toxicity of organophosphate flame retardants and polybrominated diphenyl ethers to Caenorhabditis elegans. Toxicol Sci 154, 241-252
    • 10.1093/toxsci/kfw162 Belvindrah, R., Graus-Porta, D., Goebbels, S. et al. (2007). Beta1 integrins in radial glia but not in migrating neurons are essential for the formation of cell layers in the cerebral cortex. J Neurosci 27, 13854-13865
    • Behl, M., Rice, J. R., Smith, M. V. et al. (2016). Editor’s highlight: Comparative toxicity of organophosphate flame retardants and polybrominated diphenyl ethers to Caenorhabditis elegans. Toxicol Sci 154, 241-252. doi:10.1093/toxsci/kfw162 Belvindrah, R., Graus-Porta, D., Goebbels, S. et al. (2007). Beta1 integrins in radial glia but not in migrating neurons are essential for the formation of cell layers in the cerebral cortex. J Neurosci 27, 13854-13865. doi:10.1523/JNEUROSCI.4494-07.2007
    • (2016)
    • Behl, M.1    Rice, J. R.2    Smith, M. V.3
  • 27
    • 84872678533 scopus 로고    scopus 로고
    • Ziprasidone – not haloperidol – induces more de-novo neurogenesis of adult neural stem cells derived from murine hippocampus
    • Benninghoff, J., Grunze, H., Schindler, C. et al. (2013). Ziprasidone – not haloperidol – induces more de-novo neurogenesis of adult neural stem cells derived from murine hippocampus. Pharmacopsychiatry 46, 10-15. doi:10.1055/s-0032-1311607
    • (2013) Pharmacopsychiatry , vol.46 , pp. 10-15
    • Benninghoff, J.1    Grunze, H.2    Schindler, C.3
  • 28
    • 0035940787 scopus 로고    scopus 로고
    • Role of thyroid hormones in the maturation and organisation of rat barrel cortex
    • Berbel, P., Auso, E., Garcia-Velasco, J. V. et al. (2001). Role of thyroid hormones in the maturation and organisation of rat barrel cortex. Neuroscience 107, 383-394. doi:10.1016/ S0306-4522(01)00368-2
    • (2001) Neuroscience , vol.107 , pp. 383-394
    • Berbel, P.1    Auso, E.2    Garcia-Velasco, J. V.3
  • 29
    • 84990193766 scopus 로고    scopus 로고
    • Thyroid hormones in brain development and function
    • L. J. De Groot, G. Chrousos, K. Dungan et al. (eds), South Dartmouth, MA: MDText.com, Inc
    • Bernal, J. (2015). Thyroid hormones in brain development and function. In L. J. De Groot, G. Chrousos, K. Dungan et al. (eds.), Endotext. South Dartmouth, MA: MDText.com, Inc. https://www.ncbi.nlm.nih.gov/books/NBK285549/
    • (2015) Endotext
    • Bernal, J.1
  • 30
    • 84862138497 scopus 로고    scopus 로고
    • The dynamic role of bone morphogenetic proteins in neural stem cell fate and maturation
    • Bond, A. M., Bhalala, O. G. and Kessler, J. A. (2012). The dynamic role of bone morphogenetic proteins in neural stem cell fate and maturation. Dev Neurobiol 72, 1068-1084. doi:10.1002/dneu.22022
    • (2012) Dev Neurobiol , vol.72 , pp. 1068-1084
    • Bond, A. M.1    Bhalala, O. G.2    Kessler, J. A.3
  • 31
    • 84895939484 scopus 로고    scopus 로고
    • Role of radial glial cells in cerebral cortex folding
    • Borrell, V. and Götz, M. (2014). Role of radial glial cells in cerebral cortex folding. Curr Opin Neurobiol 27, 39-46. doi:10.1016/j.conb.2014.02.007
    • (2014) Curr Opin Neurobiol , vol.27 , pp. 39-46
    • Borrell, V.1    Götz, M.2
  • 32
    • 77649192930 scopus 로고    scopus 로고
    • Oligodendrocytes: Biology and pathology
    • Bradl, M. and Lassmann, H. (2010). Oligodendrocytes: Biology and pathology. Acta Neuropathol 119, 37-53. doi:10.1007/ s00401-009-0601-5
    • (2010) Acta Neuropathol , vol.119 , pp. 37-53
    • Bradl, M.1    Lassmann, H.2
  • 33
    • 77955465057 scopus 로고    scopus 로고
    • Bio-markers of glial cell proliferation and differentiation in culture
    • Bramanti, V., Tomassoni, D., Avitabile, M. et al. (2010). Bio-markers of glial cell proliferation and differentiation in culture. Front Biosci (Schol Ed) 2, 558-570. doi:10.2741/85
    • (2010) Front Biosci (Schol Ed) , vol.2 , pp. 558-570
    • Bramanti, V.1    Tomassoni, D.2    Avitabile, M.3
  • 34
    • 49249113004 scopus 로고    scopus 로고
    • Development of a high-throughput screening assay for chemical effects on proliferation and viability of immortalized human neural progenitor cells
    • Breier, J. M., Radio, N. M., Mundy, W. R. and Shafer, T. J. (2008). Development of a high-throughput screening assay for chemical effects on proliferation and viability of immortalized human neural progenitor cells. Toxicol Sci 105, 119-133. doi:10.1093/toxsci/kfn115
    • (2008) Toxicol Sci , vol.105 , pp. 119-133
    • Breier, J. M.1    Radio, N. M.2    Mundy, W. R.3    Shafer, T. J.4
  • 35
    • 84886293335 scopus 로고    scopus 로고
    • The ontology of craniofacial development and malformation for translational craniofacial research
    • Brinkley, J. F., Borromeo, C., Clarkson, M. et al. (2013). The ontology of craniofacial development and malformation for translational craniofacial research. Am J Med Genet C Semin Med Genet 163C, 232-245. doi:10.1002/ajmg.c.31377
    • (2013) Am J Med Genet C Semin Med Genet , vol.163C , pp. 232-245
    • Brinkley, J. F.1    Borromeo, C.2    Clarkson, M.3
  • 36
    • 85006263555 scopus 로고    scopus 로고
    • Evaluation of a microelectrode array-based assay for neural network ontogeny using training set chemicals
    • Brown, J. P., Hall, D., Frank, C. L. et al. (2016). Evaluation of a microelectrode array-based assay for neural network ontogeny using training set chemicals. Toxicol Sci 154, 126-139. doi:10.1093/toxsci/kfw147
    • (2016) Toxicol Sci , vol.154 , pp. 126-139
    • Brown, J. P.1    Hall, D.2    Frank, C. L.3
  • 37
    • 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
  • 38
    • 85027501917 scopus 로고    scopus 로고
    • Correction to screening chemicals for estrogen receptor bioactivity using a computational model
    • Browne, P., Judson, R. S., Casey, W. et al. (2017). Correction to screening chemicals for estrogen receptor bioactivity using a computational model. Environ Sci Technol 51, 9415-9415. doi:10.1021/acs.est.7b03317
    • (2017) Environ Sci Technol , vol.51 , pp. 9415-9415
    • Browne, P.1    Judson, R. S.2    Casey, W.3
  • 39
    • 84859264960 scopus 로고    scopus 로고
    • Insight into the neuroproteomics effects of the food-contaminant non-dioxin like polychlorinated biphenyls
    • Brunelli, L., Llansola, M., Felipo, V. et al. (2012). Insight into the neuroproteomics effects of the food-contaminant non-dioxin like polychlorinated biphenyls. J Proteomics 75, 2417-2430. doi:10.1016/j.jprot.2012.02.023
    • (2012) J Proteomics , vol.75 , pp. 2417-2430
    • Brunelli, L.1    Llansola, M.2    Felipo, V.3
  • 40
    • 0037051159 scopus 로고    scopus 로고
    • Arsenic induced changes in growth development and apoptosis in neonatal and adult brain cells in vivo and in tissue culture
    • Chattopadhyay, S., Bhaumik, S., Nag Chaudhury, A. and Das Gupta, S. (2002). Arsenic induced changes in growth development and apoptosis in neonatal and adult brain cells in vivo and in tissue culture. Toxicol Lett 128, 73-84. doi:10.1016/ S0378-4274(01)00535-5
    • (2002) Toxicol Lett , vol.128 , pp. 73-84
    • Chattopadhyay, S.1    Bhaumik, S.2    Nag Chaudhury, A.3    Das Gupta, S.4
  • 41
    • 84867759372 scopus 로고    scopus 로고
    • Developmental lead acetate exposure induces embryonic toxicity and memory deficit in adult zebrafish
    • Chen, J., Chen, Y., Liu, W. et al. (2012a). Developmental lead acetate exposure induces embryonic toxicity and memory deficit in adult zebrafish. Neurotoxicol Teratol 34, 581-586. doi:10.1016/j.ntt.2012.09.001
    • (2012) Neurotoxicol Teratol , vol.34 , pp. 581-586
    • Chen, J.1    Chen, Y.2    Liu, W.3
  • 42
    • 84860991226 scopus 로고    scopus 로고
    • BDE-47 disrupts axonal growth and motor behavior in developing zebrafish
    • Chen, X., Huang, C., Wang, X. et al. (2012b). BDE-47 disrupts axonal growth and motor behavior in developing zebrafish. Aquat Toxicol 120-121, 35-44. doi:10.1016/j. aquatox.2012.04.014
    • (2012) Aquat Toxicol , vol.120-121 , pp. 35-44
    • Chen, X.1    Huang, C.2    Wang, X.3
  • 43
    • 0024540448 scopus 로고
    • The effects of methylmercury on the developing brain
    • Choi, B. H. (1989). The effects of methylmercury on the developing brain. Prog Neurobiol 32, 447-470. doi:10.1016/0301-0082(89)90018-X
    • (1989) Prog Neurobiol , vol.32 , pp. 447-470
    • Choi, B. H.1
  • 44
    • 84912064761 scopus 로고    scopus 로고
    • A three-dimensional human neural cell culture model of Alzheimer’s disease
    • Choi, S. H., Kim, Y. H., Hebisch, M. et al. (2014). A three-dimensional human neural cell culture model of Alzheimer’s disease. Nature 515, 274-278. doi:10.1038/nature13800
    • (2014) Nature , vol.515 , pp. 274-278
    • Choi, S. H.1    Kim, Y. H.2    Hebisch, M.3
  • 46
    • 41549164242 scopus 로고    scopus 로고
    • Cadmium inhibits neurogenesis in zebrafish embryonic brain development
    • Mol Neurodegener 11, 157
    • Mol Neurodegener 11, 75. doi:10.1186/s13024-016-0139-7 Chow, E. S., Hui, M. N., Lin, C. C. and Cheng, S. H. (2008). Cadmium inhibits neurogenesis in zebrafish embryonic brain development. Aquat Toxicol 87, 157-169. doi:10.1016/j.aquatox.2008.01.019
    • (2008) Aquat Toxicol , vol.87 , pp. 75-169
    • Chow, E. S.1    Hui, M. N.2    Lin, C. C.3    Cheng, S. H.4
  • 47
    • 0034177780 scopus 로고    scopus 로고
    • Testing methods for developmental neurotoxicity of environmental chemicals
    • Claudio, L., Kwa, W. C., Russell, A. L. and Wallinga, D. (2000). Testing methods for developmental neurotoxicity of environmental chemicals. Toxicol Appl Pharmacol 164, 1-14. doi:10.1006/taap.2000.8890
    • (2000) Toxicol Appl Pharmacol , vol.164 , pp. 1-14
    • Claudio, L.1    Kwa, W. C.2    Russell, A. L.3    Wallinga, D.4
  • 48
    • 34250191475 scopus 로고    scopus 로고
    • Workgroup report: Incorporating in vitro alternative methods for developmental neurotoxicity into international hazard and risk assessment strategies
    • Coecke, S., Goldberg, A. M., Allen, S. et al. (2007). Workgroup report: Incorporating in vitro alternative methods for developmental neurotoxicity into international hazard and risk assessment strategies. Environ Health Perspect 115, 924-931. doi:10.1289/ehp.9427
    • (2007) Environ Health Perspect , vol.115 , pp. 924-931
    • Coecke, S.1    Goldberg, A. M.2    Allen, S.3
  • 49
    • 0025159354 scopus 로고
    • The central nervous system in the Apert syndrome
    • Cohen, M. M. Jr. and Kreiborg, S. (1990). The central nervous system in the Apert syndrome. Am J Med Genet 35, 36-45. doi:10.1002/ajmg.1320350108
    • (1990) Am J Med Genet , vol.35 , pp. 36-45
    • Cohen, M. M.1    Kreiborg, S.2
  • 50
    • 55849130994 scopus 로고    scopus 로고
    • Communication between the synapse and the nucleus in neuronal development, plasticity and disease
    • Cohen, S. and Greenberg, M. E. (2008). Communication between the synapse and the nucleus in neuronal development, plasticity and disease. Annu Rev Cell Dev Biol 24, 183-209. doi:10.1146/annurev.cellbio.24.110707.175235
    • (2008) Annu Rev Cell Dev Biol , vol.24 , pp. 183-209
    • Cohen, S.1    Greenberg, M. E.2
  • 51
    • 81855211078 scopus 로고    scopus 로고
    • Development of a neural teratogenicity test based on human embryonic stem cells: Response to retinoic acid exposure
    • Colleoni, S., Galli, C., Gaspar, J. A. et al. (2011). Development of a neural teratogenicity test based on human embryonic stem cells: Response to retinoic acid exposure. Toxicol Sci 124, 370-377. doi:10.1093/toxsci/kfr245
    • (2011) Toxicol Sci , vol.124 , pp. 370-377
    • Colleoni, S.1    Galli, C.2    Gaspar, J. A.3
  • 52
    • 84871327000 scopus 로고    scopus 로고
    • Characterisation of a neural teratogenicity assay based on human ESCs differentiation following exposure to valproic acid
    • Colleoni, S., Galli, C., Gaspar, J. A. et al. (2012). Characterisation of a neural teratogenicity assay based on human ESCs differentiation following exposure to valproic acid. Curr Med Chem 19, 6065-6071. doi:10.2174/0929867311209066065
    • (2012) Curr Med Chem , vol.19 , pp. 6065-6071
    • Colleoni, S.1    Galli, C.2    Gaspar, J. A.3
  • 53
    • 0034081061 scopus 로고    scopus 로고
    • Acetylcholine induces neuritic outgrowth in rat primary olfactory bulb cultures
    • Coronas, V., Durand, M., Chabot, J. G. et al. (2000). Acetylcholine induces neuritic outgrowth in rat primary olfactory bulb cultures. Neuroscience 98, 213-219. doi:10.1016/S0306-4522(00)00143-3
    • (2000) Neuroscience , vol.98 , pp. 213-219
    • Coronas, V.1    Durand, M.2    Chabot, J. G.3
  • 54
    • 84886826594 scopus 로고    scopus 로고
    • mTOR complexes in neurodevelopmental and neuropsychiatric disorders
    • Costa-Mattioli, M. and Monteggia, L. M. (2013). mTOR complexes in neurodevelopmental and neuropsychiatric disorders. Nat Neurosci 16, 1537-1543. doi:10.1038/nn.3546
    • (2013) Nat Neurosci , vol.16 , pp. 1537-1543
    • Costa-Mattioli, M.1    Monteggia, L. M.2
  • 55
    • 84970990772 scopus 로고    scopus 로고
    • Characterization of early cortical neural network development in multiwell microelectrode array plates
    • Cotterill, E., Hall, D., Wallace, K. et al. (2016). Characterization of early cortical neural network development in multiwell microelectrode array plates. J Biomol Screen 21, 510-519. doi:10.1177/1087057116640520
    • (2016) J Biomol Screen , vol.21 , pp. 510-519
    • Cotterill, E.1    Hall, D.2    Wallace, K.3
  • 56
    • 84858442380 scopus 로고    scopus 로고
    • Developmental exposure to valproate and ethanol alters locomotor activity and retino-tectal projection area in zebrafish embryos
    • Cowden, J., Padnos, B., Hunter, D. et al. (2012). Developmental exposure to valproate and ethanol alters locomotor activity and retino-tectal projection area in zebrafish embryos. Reprod Toxicol 33, 165-173. doi:10.1016/j.reprotox.2011.11.111
    • (2012) Reprod Toxicol , vol.33 , pp. 165-173
    • Cowden, J.1    Padnos, B.2    Hunter, D.3
  • 57
    • 2042441794 scopus 로고    scopus 로고
    • A qualitative retrospective analysis of positive control data in developmental neurotoxicity studies
    • Crofton, K. M., Makris, S. L., Sette, W. F. et al. (2004). A qualitative retrospective analysis of positive control data in developmental neurotoxicity studies. Neurotoxicol Teratol 26, 345-352. doi:10.1016/j.ntt.2004.02.007
    • (2004) Neurotoxicol Teratol , vol.26 , pp. 345-352
    • Crofton, K. M.1    Makris, S. L.2    Sette, W. F.3
  • 58
    • 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
  • 59
    • 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
  • 60
    • 0033955358 scopus 로고    scopus 로고
    • Developmental neurotoxicity of chlorpyrifos in vivo and in vitro: Effects on nuclear transcription factors involved in cell replication and differentiation
    • Crumpton, T. L., Seidler, F. J. and Slotkin, T. A. (2000). Developmental neurotoxicity of chlorpyrifos in vivo and in vitro: Effects on nuclear transcription factors involved in cell replication and differentiation. Brain Res 857, 87-98. doi:10.1016/ S0006-8993(99)02357-4
    • (2000) Brain Res , vol.857 , pp. 87-98
    • Crumpton, T. L.1    Seidler, F. J.2    Slotkin, T. A.3
  • 61
    • 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
  • 62
    • 57349130542 scopus 로고    scopus 로고
    • Minireview: Expanding the mind: Insulin-like growth factor I and brain development
    • D’Ercole, A. J. and Ye, P. (2008). Minireview: Expanding the mind: Insulin-like growth factor I and brain development. Endocrinology 149, 5958-5962. doi:10.1210/en.2008-0920
    • (2008) Endocrinology , vol.149 , pp. 5958-5962
    • D’Ercole, A. J.1    Ye, P.2
  • 63
    • 84864024084 scopus 로고    scopus 로고
    • Dopaminergic neurons from midbrain-specified human embryonic stem ceel-derived neural stem cells engrafted in a monkey model of Parkinson’s disease
    • Daadi, M. M., Grueter, B. A., Malenka, R. C. et al. (2012). Dopaminergic neurons from midbrain-specified human embryonic stem ceel-derived neural stem cells engrafted in a monkey model of Parkinson’s disease. PLoS One 7, e41120. doi:10.1371/journal.pone.0041120
    • (2012) PLoS One , vol.7 , pp. e41120
    • Daadi, M. M.1    Grueter, B. A.2    Malenka, R. C.3
  • 64
    • 85015747832 scopus 로고    scopus 로고
    • BDE-99 impairs differentiation of human and mouse NPCs into the oligodendroglial lineage by species-specific modes of action
    • Dach, K., Bendt, F., Huebenthal, U. et al. (2017). BDE-99 impairs differentiation of human and mouse NPCs into the oligodendroglial lineage by species-specific modes of action. Sci Rep 7, 44861. doi:10.1038/srep44861
    • (2017) Sci Rep , vol.7 , pp. 44861
    • Dach, K.1    Bendt, F.2    Huebenthal, U.3
  • 65
    • 84946721769 scopus 로고    scopus 로고
    • Animal use for science in Europe
    • Daneshian, M., Busquet, F., Hartung, T. et al. (2015). Animal use for science in Europe. ALTEX 32, 261-274. doi:10.14573/ altex.1509081
    • (2015) ALTEX , vol.32 , pp. 261-274
    • Daneshian, M.1    Busquet, F.2    Hartung, T.3
  • 66
    • 84961653980 scopus 로고    scopus 로고
    • Launch of a large integrated European in vitro toxicology project: EU-ToxRisk
    • Daneshian, M., Kamp, H., Hengstler, J. et al. (2016). 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
  • 67
    • 84971557656 scopus 로고    scopus 로고
    • Zika virus depletes neural progenitors in human cerebral organoids through activation of the innate immune receptor TLR3
    • Dang, J., Tiwari, S. K., Lichinchi, G. et al. (2016). Zika virus depletes neural progenitors in human cerebral organoids through activation of the innate immune receptor TLR3. Cell Stem Cell 19, 258-265. doi:10.1016/j.stem.2016.04.014
    • (2016) Cell Stem Cell , vol.19 , pp. 258-265
    • Dang, J.1    Tiwari, S. K.2    Lichinchi, G.3
  • 68
    • 3042538236 scopus 로고    scopus 로고
    • Maternal thyroid hormones early in pregnancy and fetal brain development
    • de Escobar, G. M., Obregon, M. J. and del Rey, F. E. (2004). Maternal thyroid hormones early in pregnancy and fetal brain development. Best Pract Res Clin Endocrinol Metab 18, 225-248. doi:10.1016/j.beem.2004.03.012
    • (2004) Best Pract Res Clin Endocrinol Metab , vol.18 , pp. 225-248
    • de Escobar, G. M.1    Obregon, M. J.2    del Rey, F. E.3
  • 69
    • 85049303762 scopus 로고    scopus 로고
    • A high-throughput approach to identify specific neurotoxicants / developmental toxicants in human neuronal cell function assays
    • Delp, J., Gutbier, S., Klima, S. et al. (2018). A high-throughput approach to identify specific neurotoxicants / developmental toxicants in human neuronal cell function assays. ALTEX 35, 235-253. doi:10.14573/altex.1712182
    • (2018) ALTEX , vol.35 , pp. 235-253
    • Delp, J.1    Gutbier, S.2    Klima, S.3
  • 70
    • 84994442740 scopus 로고    scopus 로고
    • The adverse outcome pathway concept: A basis for developing regulatory decision-making tools
    • Delrue, N., Sachana, M., Sakuratani, Y. et al. (2016). The adverse outcome pathway concept: A basis for developing regulatory decision-making tools. Altern Lab Anim 44, 417-429.
    • (2016) Altern Lab Anim , vol.44 , pp. 417-429
    • Delrue, N.1    Sachana, M.2    Sakuratani, Y.3
  • 71
    • 85028346501 scopus 로고    scopus 로고
    • Infectious causes of microcephaly: Epidemiology, pathogenesis, diagnosis, and management
    • Devakumar, D., Bamford, A., Ferreira, M. U. et al. (2018). Infectious causes of microcephaly: Epidemiology, pathogenesis, diagnosis, and management. Lancet Infect Dis 18, e1-e13. doi:10.1016/S1473-3099(17)30398-5
    • (2018) Lancet Infect Dis , vol.18 , pp. e1-e13
    • Devakumar, D.1    Bamford, A.2    Ferreira, M. U.3
  • 72
    • 84991201245 scopus 로고    scopus 로고
    • Chronic 14-day exposure to insecticides or methylmercury modulates neuronal activity in primary rat cortical cultures
    • Dingemans, M. M., Schütte, M. G., Wiersma, D. M. et al. (2016). Chronic 14-day exposure to insecticides or methylmercury modulates neuronal activity in primary rat cortical cultures. Neurotoxicology 57, 194-202. doi:10.1016/j.neuro.2016.10.002
    • (2016) Neurotoxicology , vol.57 , pp. 194-202
    • Dingemans, M. M.1    Schütte, M. G.2    Wiersma, D. M.3
  • 73
    • 84860393640 scopus 로고    scopus 로고
    • Effects of lead on neurogenesis during zebrafish embryonic brain development
    • Dou, C. and Zhang, J. (2011). Effects of lead on neurogenesis during zebrafish embryonic brain development. J Hazard Mater 194, 277-282. doi:10.1016/j.jhazmat.2011.07.106
    • (2011) J Hazard Mater , vol.194 , pp. 277-282
    • Dou, C.1    Zhang, J.2
  • 74
    • 84939235734 scopus 로고    scopus 로고
    • Grouping of histone deacetylase inhibitors and other toxicants disturbing neural crest migration by transcriptional profiling
    • Dreser, N., Zimmer, B., Dietz, C. et al. (2015). Grouping of histone deacetylase inhibitors and other toxicants disturbing neural crest migration by transcriptional profiling. Neurotoxicology 50, 56-70. doi:10.1016/j.neuro.2015.07.008
    • (2015) Neurotoxicology , vol.50 , pp. 56-70
    • Dreser, N.1    Zimmer, B.2    Dietz, C.3
  • 75
    • 84927722477 scopus 로고    scopus 로고
    • Sensitivity of neuroprogenitor cells to chemical-induced apoptosis using a multiplexed assay suitable for high-throughput screening
    • Druwe, I., Freudenrich, T. M., Wallace, K. et al. (2015). Sensitivity of neuroprogenitor cells to chemical-induced apoptosis using a multiplexed assay suitable for high-throughput screening. Toxicology 333, 14-24. doi:10.1016/j.tox.2015.03.011
    • (2015) Toxicology , vol.333 , pp. 14-24
    • Druwe, I.1    Freudenrich, T. M.2    Wallace, K.3
  • 76
    • 85051031270 scopus 로고    scopus 로고
    • Comparison of human induced pluripotent stem cell-derived neurons and rat primary cortical neurons as in vitro models of neurite outgrowth
    • Druwe, I., Freudenrich, T. M., Wallace, K. et al. (2016). Comparison of human induced pluripotent stem cell-derived neurons and rat primary cortical neurons as in vitro models of neurite outgrowth. Appl In Vitro Toxicol 2, 26-36. doi:10.1089/ aivt.2015.0025
    • (2016) Appl In Vitro Toxicol , vol.2 , pp. 26-36
    • Druwe, I.1    Freudenrich, T. M.2    Wallace, K.3
  • 77
    • 84860834105 scopus 로고    scopus 로고
    • Neural crest progenitors and stem cells: From early development to adulthood
    • Dupin, E. and Sommer, L. (2012). Neural crest progenitors and stem cells: From early development to adulthood. Dev Biol 366, 83-95. doi:10.1016/j.ydbio.2012.02.035
    • (2012) Dev Biol , vol.366 , pp. 83-95
    • Dupin, E.1    Sommer, L.2
  • 78
    • 20044382806 scopus 로고    scopus 로고
    • Role for Akt3/ protein kinase bγ in attainment of normal brain size
    • Easton, R. M., Cho, H., Roovers, K. et al. (2005). Role for Akt3/ protein kinase bγ in attainment of normal brain size. Mol Cell Biol 25, 1869-1878. doi:10.1128/MCB.25.5.1869-1878.2005
    • (2005) Mol Cell Biol , vol.25 , pp. 1869-1878
    • Easton, R. M.1    Cho, H.2    Roovers, K.3
  • 79
    • 85026458064 scopus 로고    scopus 로고
    • Gestational age and sex influence the susceptibility of human neural progenitor cells to low levels of MeHg
    • Edoff, K., Raciti, M., Moors, M. et al. (2017). Gestational age and sex influence the susceptibility of human neural progenitor cells to low levels of MeHg. Neurotox Res 32, 683-693. doi:10.1007/s12640-017-9786-x
    • (2017) Neurotox Res , vol.32 , pp. 683-693
    • Edoff, K.1    Raciti, M.2    Moors, M.3
  • 80
    • 84937513010 scopus 로고    scopus 로고
    • Prevention of human dopaminergic neurodegeneration in an astrocytes co-culture system allowing endogenous drug metabolism
    • Efremova, L., Schildknecht, S., Adam, M. et al. (2015). Prevention of human dopaminergic neurodegeneration in an astrocytes co-culture system allowing endogenous drug metabolism. Br J Pharmacol 172, 4119-4132. doi:10.1111/bph.13193
    • (2015) Br J Pharmacol , vol.172 , pp. 4119-4132
    • Efremova, L.1    Schildknecht, S.2    Adam, M.3
  • 81
    • 84962711474 scopus 로고    scopus 로고
    • Switching from astrocytic neuroprotection to neurodegeneration by cytokine stimulation
    • Efremova, L., Chovancova, P., Adam, M. et al. (2017). Switching from astrocytic neuroprotection to neurodegeneration by cytokine stimulation. Arch Toxicol 91, 231-246. doi:10.1007/ s00204-016-1702-2
    • (2017) Arch Toxicol , vol.91 , pp. 231-246
    • Efremova, L.1    Chovancova, P.2    Adam, M.3
  • 82
    • 85020073858 scopus 로고    scopus 로고
    • Scientific opinion on the developmental neurotoxicity potential of acetamiprid and imidacloprid
    • EFSA Panel on Plant Protection Products and their Residues. (2013). Scientific opinion on the developmental neurotoxicity potential of acetamiprid and imidacloprid. EFSA Journal 11, 3471. doi:10.2903/j.efsa.2013.3471
    • (2013) EFSA Journal , vol.11 , pp. 3471
  • 83
    • 84954401234 scopus 로고    scopus 로고
    • Plasticity-related genes in brain development and amygdala-dependent learning
    • Ehrlich, D. E. and Josselyn, S. A. (2016). Plasticity-related genes in brain development and amygdala-dependent learning. Genes Brain Behav 15, 125-143. doi:10.1111/gbb.12255
    • (2016) Genes Brain Behav , vol.15 , pp. 125-143
    • Ehrlich, D. E.1    Josselyn, S. A.2
  • 85
    • 84960867203 scopus 로고    scopus 로고
    • Should the scope of human mixture risk assessment span legislative/regulatory silos for chemicals?
    • Evans, R. M., Martin, O. V., Faust, M. and Kortenkamp, A. (2016). Should the scope of human mixture risk assessment span legislative/regulatory silos for chemicals? Sci Total Environ 543, 757-764. doi:10.1016/j.scitotenv.2015.10.162
    • (2016) Sci Total Environ , vol.543 , pp. 757-764
    • Evans, R. M.1    Martin, O. V.2    Faust, M.3    Kortenkamp, A.4
  • 86
    • 18744404492 scopus 로고    scopus 로고
    • Overgrowth and trisomy 15q26.1-qter including the IGF1 receptor gene: Report of two families and review of the literature
    • Faivre, L., Gosset, P., Cormier-Daire, V. et al. (2002). Overgrowth and trisomy 15q26.1-qter including the IGF1 receptor gene: Report of two families and review of the literature. Eur J Hum Genet 10, 699-706. doi:10.1038/sj.ejhg.5200879
    • (2002) Eur J Hum Genet , vol.10 , pp. 699-706
    • Faivre, L.1    Gosset, P.2    Cormier-Daire, V.3
  • 87
    • 33645099728 scopus 로고    scopus 로고
    • The inflammatory transcriptome of reactive murine astrocytes and implications for their innate immune functions
    • Falsig, J., Porzgen, P., Lund, S. et al. (2006). The inflammatory transcriptome of reactive murine astrocytes and implications for their innate immune functions. J Neurochem 96, 893-907. doi:10.1111/j.1471-4159.2005.03622.x
    • (2006) J Neurochem , vol.96 , pp. 893-907
    • Falsig, J.1    Porzgen, P.2    Lund, S.3
  • 88
    • 34249661591 scopus 로고    scopus 로고
    • Phosphorylation of beta-catenin by AKT promotes beta-catenin transcriptional activity
    • Fang, D., Hawke, D., Zheng, Y. et al. (2007). Phosphorylation of beta-catenin by AKT promotes beta-catenin transcriptional activity. J Biol Chem 282, 11221-11229. doi:10.1074/jbc. M611871200
    • (2007) J Biol Chem , vol.282 , pp. 11221-11229
    • Fang, D.1    Hawke, D.2    Zheng, Y.3
  • 89
    • 85018480249 scopus 로고    scopus 로고
    • Application of IATA – A case study in evaluating the global and local performance of a Bayesian network model for skin sensitization
    • Fitzpatrick, J. M. and Patlewicz, G. (2017). Application of IATA – A case study in evaluating the global and local performance of a Bayesian network model for skin sensitization. SAR QSAR Environ Res 28, 297-310. doi:10.1080/106293 6X.2017.1311941
    • (2017) SAR QSAR Environ Res , vol.28 , pp. 297-310
    • Fitzpatrick, J. M.1    Patlewicz, G.2
  • 90
    • 84885739130 scopus 로고    scopus 로고
    • Functional characterisation of the maturation of the blood-brain barrier in larval zebrafish
    • Fleming, A., Diekmann, H. and Goldsmith, P. (2013). Functional characterisation of the maturation of the blood-brain barrier in larval zebrafish. PLoS One 8, e77548. doi:10.1371/journal. pone.0077548
    • (2013) PLoS One , vol.8 , pp. e77548
    • Fleming, A.1    Diekmann, H.2    Goldsmith, P.3
  • 91
    • 0346882728 scopus 로고    scopus 로고
    • Hemimegalencephaly: Part 2. Neuropathology suggests a disorder of cellular lineage
    • Flores-Sarnat, L., Sarnat, H. B., Dávila-Gutiérrez, G. and Alvarez, A. (2003). Hemimegalencephaly: Part 2. Neuropathology suggests a disorder of cellular lineage. J Child Neurol 18, 776-785. doi:10.1177/08830738030180111101
    • (2003) J Child Neurol , vol.18 , pp. 776-785
    • Flores-Sarnat, L.1    Sarnat, H. B.2    Dávila-Gutiérrez, G.3    Alvarez, A.4
  • 92
    • 84880039835 scopus 로고    scopus 로고
    • HDAC inhibitors dysregulate neural stem cell activity in the postnatal mouse brain
    • Foti, S. B., Chou, A., Moll, A. D. and Roskams, A. J. (2013). HDAC inhibitors dysregulate neural stem cell activity in the postnatal mouse brain. Int J Dev Neurosci 31, 434-447. doi:10.1016/j.ijdevneu.2013.03.008
    • (2013) Int J Dev Neurosci , vol.31 , pp. 434-447
    • Foti, S. B.1    Chou, A.2    Moll, A. D.3    Roskams, A. J.4
  • 93
    • 85038023243 scopus 로고    scopus 로고
    • Developmental neurotoxicants disrupt formation of cortical networks on microelectrode arrays: Screening 86 compounds in the neural network formation assay
    • Frank, C. L., Brown, J. P., Wallace, K. et al. (2017). Developmental neurotoxicants disrupt formation of cortical networks on microelectrode arrays: Screening 86 compounds in the neural network formation assay. Toxicol Sci 160, 121-135. doi:10.1093/toxsci/kfx169
    • (2017) Toxicol Sci , vol.160 , pp. 121-135
    • Frank, C. L.1    Brown, J. P.2    Wallace, K.3
  • 94
    • 7444264727 scopus 로고    scopus 로고
    • Pten loss causes hypertrophy and increased proliferation of astrocytes in vivo
    • Fraser, M. M., Zhu, X., Kwon, C. H. et al. (2004). Pten loss causes hypertrophy and increased proliferation of astrocytes in vivo. Cancer Res 64, 7773-7779. doi:10.1158/0008-5472. CAN-04-2487
    • (2004) Cancer Res , vol.64 , pp. 7773-7779
    • Fraser, M. M.1    Zhu, X.2    Kwon, C. H.3
  • 95
    • 85032719658 scopus 로고    scopus 로고
    • Literature review on in vitro and alternative developmental neurotoxicity (DNT) testing methods
    • Fritsche, E., Alm, H., Baumann, J. et al. (2015). Literature review on in vitro and alternative developmental neurotoxicity (DNT) testing methods. EFSA Supporting Publications 12, 4. doi:10.2903/sp.efsa.2015.EN-778
    • (2015) EFSA Supporting Publications , vol.12 , pp. 4
    • Fritsche, E.1    Alm, H.2    Baumann, J.3
  • 96
    • 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. (2017a). 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
  • 99
    • 0030601994 scopus 로고    scopus 로고
    • T-cell stimulus-induced crosstalk between lymphocytes and liver cells results in augmented cytokine-release
    • Gantner, F., Leist, M., Küsters, S. et al. (1996). T-cell stimulus-induced crosstalk between lymphocytes and liver cells results in augmented cytokine-release. Exp Cell Res 229, 137-146. doi:10.1006/excr.1996.0351
    • (1996) Exp Cell Res , vol.229 , pp. 137-146
    • Gantner, F.1    Leist, M.2    Küsters, S.3
  • 101
    • 78149315835 scopus 로고    scopus 로고
    • Species-specific differential AhR expression protects human neural progenitor cells against developmental neurotoxicity of PAHs
    • Gassmann, K., Abel, J., Bothe, H. et al. (2010). Species-specific differential AhR expression protects human neural progenitor cells against developmental neurotoxicity of PAHs. Environ Health Perspect 118, 1571-1577. doi:10.1289/ehp.0901545
    • (2010) Environ Health Perspect , vol.118 , pp. 1571-1577
    • Gassmann, K.1    Abel, J.2    Bothe, H.3
  • 102
    • 84863835842 scopus 로고    scopus 로고
    • Automated neurosphere sorting and plating by the COPAS large particle sorter is a suitable method for high-throughput 3D in vitroapplications
    • Gassmann, K., Baumann, J., Giersiefer, S. et al. (2012). Automated neurosphere sorting and plating by the COPAS large particle sorter is a suitable method for high-throughput 3D in vitroapplications.ToxicolInVitro26,993-1000.doi:10.1016/j.tiv.2012.04.025
    • (2012) ToxicolInVitro26 , pp. 993-1000
    • Gassmann, K.1    Baumann, J.2    Giersiefer, S.3
  • 103
    • 0035003589 scopus 로고    scopus 로고
    • Cascade of caspase activation in potassium-deprived cerebellar granule neurons: Targets for treatment with peptide and protein inhibitors of apoptosis
    • Gerhardt, E., Kügler, S., Leist, M. et al. (2001). Cascade of caspase activation in potassium-deprived cerebellar granule neurons: Targets for treatment with peptide and protein inhibitors of apoptosis. Mol Cell Neurosci 17, 717-731. doi:10.1006/ mcne.2001.0962
    • (2001) Mol Cell Neurosci , vol.17 , pp. 717-731
    • Gerhardt, E.1    Kügler, S.2    Leist, M.3
  • 104
    • 0035129665 scopus 로고    scopus 로고
    • Prenatal exposure to methylmercury changes dopamine-modulated motor activity during early ontogeny: Age and gender-dependent effects
    • Gimenez-Llort, L., Ahlbom, E., Dare, E. et al. (2001). Prenatal exposure to methylmercury changes dopamine-modulated motor activity during early ontogeny: Age and gender-dependent effects. Environ Toxicol Pharmacol 9, 61-70. doi:10.1016/ S1382-6689(00)00060-0
    • (2001) Environ Toxicol Pharmacol , vol.9 , pp. 61-70
    • Gimenez-Llort, L.1    Ahlbom, E.2    Dare, E.3
  • 105
    • 84869156976 scopus 로고    scopus 로고
    • Development and homeostasis of “resident” myeloid cells: The case of the microglia
    • Gomez Perdiguero, E., Schulz, C. and Geissmann, F. (2013). Development and homeostasis of “resident” myeloid cells: The case of the microglia. Glia 61, 112-120. doi:10.1002/glia.22393
    • (2013) Glia , vol.61 , pp. 112-120
    • Gomez Perdiguero, E.1    Schulz, C.2    Geissmann, F.3
  • 106
    • 84976907426 scopus 로고    scopus 로고
    • Non-animal models of epithelial barriers (skin, intestine and lung) in research, industrial applications and regulatory toxicology
    • Gordon, S., Daneshian, M., Bouwstra, J. et al. (2015). Non-animal models of epithelial barriers (skin, intestine and lung) in research, industrial applications and regulatory toxicology. ALTEX 32, 327-378. doi:10.14573/altex.1510051
    • (2015) ALTEX , vol.32 , pp. 327-378
    • Gordon, S.1    Daneshian, M.2    Bouwstra, J.3
  • 107
    • 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
  • 108
    • 84988815127 scopus 로고    scopus 로고
    • Direct effects of ethanol on neuronal differentiation: An in vitro analysis of viability and morphology
    • Guadagnoli, T., Caltana, L., Vacotto, M. et al. (2016). Direct effects of ethanol on neuronal differentiation: An in vitro analysis of viability and morphology. Brain Res Bull 127, 177-186. doi:10.1016/j.brainresbull.2016.09.013
    • (2016) Brain Res Bull , vol.127 , pp. 177-186
    • Guadagnoli, T.1    Caltana, L.2    Vacotto, M.3
  • 109
    • 63049132535 scopus 로고    scopus 로고
    • Cadmium modulates proliferation and differentiation of human neuroblasts
    • Gulisano, M., Pacini, S. Punzi, T. et al. (2009). Cadmium modulates proliferation and differentiation of human neuroblasts. J Neurosci Res 87, 228-237. doi:10.1002/jnr.21830
    • (2009) J Neurosci Res , vol.87 , pp. 228-237
    • Gulisano, M.1    Pacini, S.2    Punzi, T.3
  • 110
    • 84877744858 scopus 로고    scopus 로고
    • Novel application of stem cell-derived neurons to evaluate the time- and dose-dependent progression of excitotoxic injury
    • Gut, I. M., Beske, P. H., Hubbard, K. S. et al. (2013). Novel application of stem cell-derived neurons to evaluate the time- and dose-dependent progression of excitotoxic injury. PLoS One 8, e64423. doi:10.1371/journal.pone.0064423
    • (2013) PLoS One , vol.8 , pp. e64423
    • Gut, I. M.1    Beske, P. H.2    Hubbard, K. S.3
  • 111
    • 0033584469 scopus 로고    scopus 로고
    • Maternal thyroid deficiency during pregnancy and subsequent neuropsychological development of the child
    • Haddow, J. E., Palomaki, G. E., Allan, W. C. et al. (1999). Maternal thyroid deficiency during pregnancy and subsequent neuropsychological development of the child. N Engl J Med 341, 549-555. doi:10.1056/NEJM199908193410801
    • (1999) N Engl J Med , vol.341 , pp. 549-555
    • Haddow, J. E.1    Palomaki, G. E.2    Allan, W. C.3
  • 112
    • 77950076985 scopus 로고    scopus 로고
    • Neurogenic radial glia in the outer subventricular zone of human neocortex
    • Hansen, D. V., Lui, J. H., Parker, P. R. and Kriegstein, A. R. (2010). Neurogenic radial glia in the outer subventricular zone of human neocortex. Nature 464, 554-561. doi:10.1038/ nature08845
    • (2010) Nature , vol.464 , pp. 554-561
    • Hansen, D. V.1    Lui, J. H.2    Parker, P. R.3    Kriegstein, A. R.4
  • 113
    • 0033940543 scopus 로고    scopus 로고
    • Additive effects of caspase inhibitor and lazaroid on the survival of transplanted rat and human embryonic dopamine neurons
    • Hansson, O., Castilho, R. F., Kaminski Schierle, G. S. et al. (2000). Additive effects of caspase inhibitor and lazaroid on the survival of transplanted rat and human embryonic dopamine neurons. Exp Neurol 164, 102-111. doi:10.1006/ exnr.2000.7406
    • (2000) Exp Neurol , vol.164 , pp. 102-111
    • Hansson, O.1    Castilho, R. F.2    Kaminski Schierle, G. S.3
  • 114
    • 77952673694 scopus 로고    scopus 로고
    • Quantitative assessment of neurite outgrowth in human embryonic stem cell-derived hN2 cells using automated high-content image analysis
    • Harrill, J. A., Freudenrich, T. M., Machacek, D. W. et al. (2010). Quantitative assessment of neurite outgrowth in human embryonic stem cell-derived hN2 cells using automated high-content image analysis. Neurotoxicology 31, 277-290. doi:10.1016/j. neuro.2010.02.003
    • (2010) Neurotoxicology , vol.31 , pp. 277-290
    • Harrill, J. A.1    Freudenrich, T. M.2    Machacek, D. W.3
  • 115
    • 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. (2011a). 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
  • 116
    • 78650524432 scopus 로고    scopus 로고
    • Use of high content image analysis to detect chemical-induced changes in synaptogenesis in vitro
    • Harrill, J. A., Robinette, B. L. and Mundy, W. R. (2011b). Use of high content image analysis to detect chemical-induced changes in synaptogenesis in vitro. Toxicol In Vitro 25, 368-387. doi:10.1016/j.tiv.2010.10.011
    • (2011) Toxicol In Vitro , vol.25 , pp. 368-387
    • Harrill, J. A.1    Robinette, B. L.2    Mundy, W. R.3
  • 117
    • 84924171530 scopus 로고    scopus 로고
    • Ontogeny of biochemical, morphological and functional parameters of synaptogenesis in primary cultures of rat hippocampal and cortical neurons
    • Harrill, J. A., Chen, H., Streifel, K. M. et al. (2015a). Ontogeny of biochemical, morphological and functional parameters of synaptogenesis in primary cultures of rat hippocampal and cortical neurons. Mol Brain 8, 10. doi:10.1186/s13041-015-0099-9
    • (2015) Mol Brain , vol.8 , pp. 10
    • Harrill, J. A.1    Chen, H.2    Streifel, K. M.3
  • 118
    • 84930822859 scopus 로고    scopus 로고
    • Media formulation influences chemical effects on neuronal growth and morphology
    • Harrill, J. A., Robinette, B. L., Freudenrich, T. M. and Mundy, W. R. (2015b). Media formulation influences chemical effects on neuronal growth and morphology. In Vitro Cell Dev Biol Anim 51, 612-629. doi:10.1007/s11626-015-9873-3
    • (2015) Vitro Cell Dev Biol Anim , vol.51 , pp. 612-629
    • Harrill, J. A.1    Robinette, B. L.2    Freudenrich, T. M.3    Mundy, W. R.4
  • 119
    • 19944430250 scopus 로고    scopus 로고
    • A modular approach to the ECVAM principles on test validity
    • Hartung, T., Bremer, S., Casati, S. et al. (2004). A modular approach to the ECVAM principles on test validity. Altern Lab Anim 32, 467-472.
    • (2004) Altern Lab Anim , vol.32 , pp. 467-472
    • Hartung, T.1    Bremer, S.2    Casati, S.3
  • 120
    • 59849129394 scopus 로고    scopus 로고
    • Food for thought ... on the evolution of toxicology and phasing out of animal testing
    • Hartung, T. and Leist, M. (2008). Food for thought ... on the evolution of toxicology and phasing out of animal testing. ALTEX 25, 91-96. doi:10.14573/altex.2008.2.91
    • (2008) ALTEX , vol.25 , pp. 91-96
    • Hartung, T.1    Leist, M.2
  • 121
    • 85018524897 scopus 로고    scopus 로고
    • Systems toxicology II: A special issue
    • Hartung, T., Kavlock, R. and Sturla, S. J. (2017a). Systems toxicology II: A special issue. Chem Res Toxicol 30, 869. doi:10.1021/acs.chemrestox.7b00038
    • (2017) Chem Res Toxicol , vol.30 , pp. 869
    • Hartung, T.1    Kavlock, R.2    Sturla, S. J.3
  • 122
    • 85018472124 scopus 로고    scopus 로고
    • Systems toxicology: Real world applications and opportunities
    • Hartung, T., FitzGerald, R. E., Jennings, P. et al. (2017b). Systems toxicology: Real world applications and opportunities. Chem Res Toxicol 30, 870-882. doi:10.1021/acs.chemrestoz. 7b00003
    • (2017) Chem Res Toxicol , vol.30 , pp. 870-882
    • Hartung, T.1    FitzGerald, R. E.2    Jennings, P.3
  • 123
    • 84860998944 scopus 로고    scopus 로고
    • Effects of methylmercury exposure on neuronal differentiation of mouse and human embryonic stem cells
    • He, X., Imanishi, S., Sone, H. et al. (2012). Effects of methylmercury exposure on neuronal differentiation of mouse and human embryonic stem cells. Toxicol Lett 212, 1-10. doi:10.1016/j.toxlet.2012.04.011
    • (2012) Toxicol Lett , vol.212 , pp. 1-10
    • He, X.1    Imanishi, S.2    Sone, H.3
  • 124
    • 85007494517 scopus 로고    scopus 로고
    • Developmental neurotoxicity of methamidophos in the embryo-larval stages of zebrafish
    • He, X., Gao, J., Dong, T. et al. (2016). Developmental neurotoxicity of methamidophos in the embryo-larval stages of zebrafish. Int J Environ Res Public Health 14, 23. doi:10.3390/ ijerph14010023
    • (2016) Int J Environ Res Public Health , vol.14 , pp. 23
    • He, X.1    Gao, J.2    Dong, T.3
  • 125
    • 32044466838 scopus 로고    scopus 로고
    • Exploiting the PI3K/AKT pathway for cancer drug discovery
    • Hennessy, B. T., Smith, D. L., Ram, P. T. et al. (2005). Exploiting the PI3K/AKT pathway for cancer drug discovery. Nat Rev Drug Discov 4, 988-1004. doi:10.1038/nrd1902
    • (2005) Nat Rev Drug Discov , vol.4 , pp. 988-1004
    • Hennessy, B. T.1    Smith, D. L.2    Ram, P. T.3
  • 126
    • 77956580727 scopus 로고    scopus 로고
    • Maternal thyroid function during early pregnancy and cognitive functioning in early childhood: The generation R study
    • Henrichs, J., Bongers-Schokking, J. J., Schenk, J. J. et al. (2010). Maternal thyroid function during early pregnancy and cognitive functioning in early childhood: The generation R study. J Clin Endocrinol Metab 95, 4227-4234. doi:10.1210/jc.2010-0415
    • (2010) J Clin Endocrinol Metab , vol.95 , pp. 4227-4234
    • Henrichs, J.1    Bongers-Schokking, J. J.2    Schenk, J. J.3
  • 127
    • 26444491115 scopus 로고    scopus 로고
    • The cerebral cortex malformation in thanatophoric dysplasia: Neuropathology and pathogenesis
    • Hevner, R. F. (2005). The cerebral cortex malformation in thanatophoric dysplasia: Neuropathology and pathogenesis. Acta Neuropathol 110, 208-221. doi:10.1007/s00401-005-1059-8
    • (2005) Acta Neuropathol , vol.110 , pp. 208-221
    • Hevner, R. F.1
  • 128
    • 84916244126 scopus 로고    scopus 로고
    • Brain overgrowth in disorders of RTKPI3K-AKT signaling: A mosaic of malformations
    • Hevner, R. F. (2015). Brain overgrowth in disorders of RTKPI3K-AKT signaling: A mosaic of malformations. Semin Perinatol 39, 36-43. doi:10.1053/j.semperi.2014.10.006
    • (2015) Semin Perinatol , vol.39 , pp. 36-43
    • Hevner, R. F.1
  • 129
    • 84995554218 scopus 로고    scopus 로고
    • Environmental toxicology: Sensitive periods of development and neurodevelopmental disorders
    • Heyer, D. B. and Meredith, R. M. (2017). Environmental toxicology: Sensitive periods of development and neurodevelopmental disorders. Neurotoxicology 58, 23-41. doi:10.1016/j. neuro.2016.10.017
    • (2017) Neurotoxicology , vol.58 , pp. 23-41
    • Heyer, D. B.1    Meredith, R. M.2
  • 130
    • 84959346131 scopus 로고    scopus 로고
    • Multiparameter toxicity assessment of novel DOPO-derived organophosphorus flame retardants
    • Hirsch, C., Striegl, B., Mathes, S. et al. (2017). Multiparameter toxicity assessment of novel DOPO-derived organophosphorus flame retardants. Arch Toxicol 91, 407-425, doi:10.1007/ s00204-016-1680-4
    • (2017) Arch Toxicol , vol.91 , pp. 407-425
    • Hirsch, C.1    Striegl, B.2    Mathes, S.3
  • 131
    • 0034659756 scopus 로고    scopus 로고
    • Phagocytosis of non-apoptotic cells dying by caspase-independent mechanisms
    • Hirt, U., Gantner, F. and Leist, M. (2000). Phagocytosis of non-apoptotic cells dying by caspase-independent mechanisms. J Immunol 164, 6520-6529. doi:10.3390/ijms17122068
    • (2000) J Immunol , vol.164 , pp. 6520-6529
    • Hirt, U.1    Gantner, F.2    Leist, M.3
  • 132
    • 33746021377 scopus 로고    scopus 로고
    • Postnatal effect of embryonic neurogenesis disturbance on reelin level in organotypic cultures of rat hippocampus
    • Hoareau, C., Hazane, F., Le Pen, G. and Krebs, M. O. (2006). Postnatal effect of embryonic neurogenesis disturbance on reelin level in organotypic cultures of rat hippocampus. Brain Res 1097, 43-51. doi:10.1016/j.brainres.2006.04.075
    • (2006) Brain Res , vol.1097 , pp. 43-51
    • Hoareau, C.1    Hazane, F.2    Le Pen, G.3    Krebs, M. O.4
  • 134
    • 84961258170 scopus 로고    scopus 로고
    • Stem cell-derived immature human dorsal root ganglia neurons to identify peripheral neurotoxicants
    • Arch Toxicol 87, 721 487
    • Arch Toxicol 87, 721-733. doi:10.1007/s00204-012-0984-2 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. 476-733
    • Hoelting, L.1    Klima, S.2    Karreman, C.3
  • 135
    • 84855274293 scopus 로고    scopus 로고
    • Domoic acid-induced neurotoxicity is mainly mediated by the AMPA/KA Receptor: Comparison between immature and mature primary cultures of neurons and glial cells from rat cerebellum
    • Hogberg, H. T. and Bal-Price, A. K. (2011). Domoic acid-induced neurotoxicity is mainly mediated by the AMPA/KA Receptor: Comparison between immature and mature primary cultures of neurons and glial cells from rat cerebellum. J Toxicol 2011, 543512. doi:10.1155/2011/543512
    • (2011) J Toxicol , vol.2011 , pp. 543512
    • Hogberg, H. T.1    Bal-Price, A. K.2
  • 136
    • 78951483473 scopus 로고    scopus 로고
    • Application of micro-electrode arrays (MEAs) as an emerging technology for developmental neurotoxicity: Evaluation of domoic acid-induced effects in primary cultures of rat cortical neurons
    • Hogberg, H. T., Sobanski, T., Novellino, A. et al. (2011). Application of micro-electrode arrays (MEAs) as an emerging technology for developmental neurotoxicity: Evaluation of domoic acid-induced effects in primary cultures of rat cortical neurons. Neurotoxicology 32, 158-168. doi:10.1016/j.neuro.2010.10.007
    • (2011) Neurotoxicology , vol.32 , pp. 158-168
    • Hogberg, H. T.1    Sobanski, T.2    Novellino, A.3
  • 137
    • 84891357438 scopus 로고    scopus 로고
    • Toward a 3D model of human brain development for studying gene/environment interactions
    • Suppl 1
    • Hogberg, H. T., Bressler, J., Christian, K. M. et al. (2013). Toward a 3D model of human brain development for studying gene/environment interactions. Stem Cell Res Ther 4, Suppl 1, S4. doi:10.1186/scrt365
    • (2013) Stem Cell Res Ther , vol.4 , pp. S4
    • Hogberg, H. T.1    Bressler, J.2    Christian, K. M.3
  • 138
    • 84965075992 scopus 로고    scopus 로고
    • Neurotoxicity screening of (illicit) drugs using novel methods for analysis of microelectrode array (MEA) recordings
    • Hondebrink, L., Verboven, A. H. A., Drega, W. S. et al. (2016). Neurotoxicity screening of (illicit) drugs using novel methods for analysis of microelectrode array (MEA) recordings. Neurotoxicology 55, 1-9. doi:10.1016/j.neuro.2016.04.020
    • (2016) Neurotoxicology , vol.55 , pp. 1-9
    • Hondebrink, L.1    Verboven, A. H. A.2    Drega, W. S.3
  • 139
    • 85019605466 scopus 로고    scopus 로고
    • Neuropharmacological characterization of the new psychoactive substance methoxetamine
    • Hondebrink, L., Kasteel, E. E. J., Tukker, A. M. et al. (2017). Neuropharmacological characterization of the new psychoactive substance methoxetamine. Neuropharmacology 123, 1-9. doi:10.1016/j.neuropharm.2017.04.035
    • (2017) Neuropharmacology , vol.123 , pp. 1-9
    • Hondebrink, L.1    Kasteel, E. E. J.2    Tukker, A. M.3
  • 140
    • 84876798186 scopus 로고    scopus 로고
    • The zebrafish reference genome sequence and its relationship to the human genome
    • Howe, K., Clark, M. D., Torroja, C. F. et al. (2013). The zebrafish reference genome sequence and its relationship to the human genome. Nature 496, 498-503. doi:10.1038/nature12111
    • (2013) Nature , vol.496 , pp. 498-503
    • Howe, K.1    Clark, M. D.2    Torroja, C. F.3
  • 141
    • 84962288692 scopus 로고    scopus 로고
    • Heterogeneous astrocytes: Active players in CNS
    • Hu, X., Yuan, Y, Wang, D. and Su, Z. (2016). Heterogeneous astrocytes: Active players in CNS. Brain Res Bull 125, 1-18. doi:10.1016/j.brainresbull.2016.03.017
    • (2016) Brain Res Bull , vol.125 , pp. 1-18
    • Hu, X.1    Yuan, Y2    Wang, D.3    Su, Z.4
  • 142
    • 0041488911 scopus 로고    scopus 로고
    • Trk receptors: Roles in neuronal signal transduction
    • Huang, E. J. and Reichardt, L. F. (2003). Trk receptors: Roles in neuronal signal transduction. Annu Rev Biochem 72, 609-642. doi:10.1146/annurev.biochem.72.121801.161629
    • (2003) Annu Rev Biochem , vol.72 , pp. 609-642
    • Huang, E. J.1    Reichardt, L. F.2
  • 143
    • 4644305820 scopus 로고    scopus 로고
    • Induction of the neural crest and the opportunities of life on the edge
    • Huang, X. and Saint-Jeannet, J. P. (2004). Induction of the neural crest and the opportunities of life on the edge. Dev Biol 275, 1-11. doi:10.1016/j.ydbio.2004.07.033
    • (2004) Dev Biol , vol.275 , pp. 1-11
    • Huang, X.1    Saint-Jeannet, J. P.2
  • 144
    • 0037817750 scopus 로고    scopus 로고
    • Signaling at the growth cone: Ligand-receptor complexes and the control of axon growth and guidance
    • Huber, A. B., Kolodkin, A. L., Ginty, D. D. and Cloutier, J. F. (2003). Signaling at the growth cone: Ligand-receptor complexes and the control of axon growth and guidance. Annu Rev Neurosci 26, 509-563. doi:10.1146/annurev.neuro.26.010302.081139
    • (2003) Annu Rev Neurosci , vol.26 , pp. 509-563
    • Huber, A. B.1    Kolodkin, A. L.2    Ginty, D. D.3    Cloutier, J. F.4
  • 145
    • 84944531468 scopus 로고    scopus 로고
    • Dysregulation of mammalian target of rapamycin signaling in mouse models of autism
    • Huber, K. M., Klann, E., Costa-Mattioli, M. et al. (2015). Dysregulation of mammalian target of rapamycin signaling in mouse models of autism. J Neurosci 35, 13836-13842. doi:10.1523/JNEUROSCI.2656-15.2015
    • (2015) J Neurosci , vol.35 , pp. 13836-13842
    • Huber, K. M.1    Klann, E.2    Costa-Mattioli, M.3
  • 146
    • 85018500801 scopus 로고    scopus 로고
    • Computational model of secondary palate fusion and disruption
    • Hutson, M. S., Leung, M. C. K., Baker, N. C. et al. (2017). Computational model of secondary palate fusion and disruption. Chem Res Toxicol 30, 965-979. doi:10.1021/acs.chemrestox. 6b00350
    • (2017) Chem Res Toxicol , vol.30 , pp. 965-979
    • Hutson, M. S.1    Leung, M. C. K.2    Baker, N. C.3
  • 147
    • 85027587638 scopus 로고    scopus 로고
    • Lysine deacetylation by HDAC6 regulates the kinase activity of AKT in human neural progenitor cells
    • Iaconelli, J., Lalonde, J., Watmuff, B. et al. (2017). Lysine deacetylation by HDAC6 regulates the kinase activity of AKT in human neural progenitor cells. ACS Chem Biol 12, 2139-2148. doi:10.1021/acschembio.6b01014
    • (2017) ACS Chem Biol , vol.12 , pp. 2139-2148
    • Iaconelli, J.1    Lalonde, J.2    Watmuff, B.3
  • 148
    • 84945366691 scopus 로고    scopus 로고
    • Acute and developmental behavioral effects of flame retardants and related chemicals in zebrafish
    • Jarema, K. A., Hunter, D. L., Shaffer, R. M. et al. (2015). Acute and developmental behavioral effects of flame retardants and related chemicals in zebrafish. Neurotoxicol Teratol 52, 194-209. doi:10.1016/j.ntt.2015.08.010
    • (2015) Neurotoxicol Teratol , vol.52 , pp. 194-209
    • Jarema, K. A.1    Hunter, D. L.2    Shaffer, R. M.3
  • 149
    • 84961689579 scopus 로고    scopus 로고
    • The fungicide imazalil induces developmental abnormalities and alters locomotor activity during early developmental stages in zebrafish
    • Jin, Y., Zhu, Z., Wang, Y. et al. (2016). The fungicide imazalil induces developmental abnormalities and alters locomotor activity during early developmental stages in zebrafish. Chemosphere 153, 455-161. doi:10.1016/j.chemosphere.2016. 03.085
    • (2016) Chemosphere , vol.153 , pp. 455-161
    • Jin, Y.1    Zhu, Z.2    Wang, Y.3
  • 150
    • 84860308551 scopus 로고    scopus 로고
    • Compound selection for in vitro modeling of developmental neurotoxicity
    • 10.2741/4064 Kapucu, F. E., Tanskanen, J. M., Mikkonen, J. E. et al. (2012). Burst analysis tool for developing neuronal networks exhibiting highly varying action potential dynamics. Front Comput Neurosci 6, 38
    • Kadereit, S., Zimmer, B., van Thriel, C. et al. (2012). Compound selection for in vitro modeling of developmental neurotoxicity. Front Biosci (Landmark Ed) 17, 2442-2460. doi:10.2741/4064 Kapucu, F. E., Tanskanen, J. M., Mikkonen, J. E. et al. (2012). Burst analysis tool for developing neuronal networks exhibiting highly varying action potential dynamics. Front Comput Neurosci 6, 38. doi:10.3389/fncom.2012.00038
    • (2012) Front Biosci (Landmark Ed) , vol.17 , pp. 2442-2460
    • Kadereit, S.1    Zimmer, B.2    van Thriel, C.3
  • 151
    • 85017091455 scopus 로고    scopus 로고
    • Zebrafish models in neuropsychopharmacology and CNS drug discovery
    • Khan, K. M., Collier, A. D., Meshalkina, D. A. et al. (2017). Zebrafish models in neuropsychopharmacology and CNS drug discovery. Br J Pharmacol 174, 1925-1944. doi:10.1111/ bph.13754
    • (2017) Br J Pharmacol , vol.174 , pp. 1925-1944
    • Khan, K. M.1    Collier, A. D.2    Meshalkina, D. A.3
  • 152
    • 84873058635 scopus 로고    scopus 로고
    • Caffeine neuroprotects against dexamethasone-induced anxiety-like behaviour in the Zebrafish (Danio rerio)
    • Khor, Y. M., Soga, T. and Parhar, I. S. (2013). Caffeine neuroprotects against dexamethasone-induced anxiety-like behaviour in the Zebrafish (Danio rerio). Gen Comp Endocrinol 181, 310-315. doi:10.1016/j.ygcen.2012.09.021
    • (2013) Gen Comp Endocrinol , vol.181 , pp. 310-315
    • Khor, Y. M.1    Soga, T.2    Parhar, I. S.3
  • 153
    • 21744452377 scopus 로고    scopus 로고
    • Compartments and their boundaries in vertebrate brain development
    • Kiecker, C. and Lumsden, A. (2005). Compartments and their boundaries in vertebrate brain development. Nat Rev Neurosci 6, 553-564. doi:10.1038/nrn1702
    • (2005) Nat Rev Neurosci , vol.6 , pp. 553-564
    • Kiecker, C.1    Lumsden, A.2
  • 154
    • 70350558453 scopus 로고    scopus 로고
    • Identification of two distinct macrophage subsets with divergent effects causing either neurotoxicity or regeneration in the injured mouse spinal cord
    • Kigerl, K. A., Gensel, J. C., Ankeny, D. P. et al. (2009). Identification of two distinct macrophage subsets with divergent effects causing either neurotoxicity or regeneration in the injured mouse spinal cord. J Neurosci 29, 13435-13444. doi:10.1523/JNEUROSCI.3257-09.2009
    • (2009) J Neurosci , vol.29 , pp. 13435-13444
    • Kigerl, K. A.1    Gensel, J. C.2    Ankeny, D. P.3
  • 155
    • 84970967287 scopus 로고    scopus 로고
    • Healthy human CSF promotes glial differentiation of hESC-derived neural cells while retaining spontaneous activity in existing neuronal networks
    • Kiiski, H., Aänismaa, R., Tenhunen, J. et al. (2013). Healthy human CSF promotes glial differentiation of hESC-derived neural cells while retaining spontaneous activity in existing neuronal networks. Biol Open 2, 605-612. doi:10.1242/ bio.20134648
    • (2013) Biol Open , vol.2 , pp. 605-612
    • Kiiski, H.1    Aänismaa, R.2    Tenhunen, J.3
  • 156
    • 84883385441 scopus 로고    scopus 로고
    • Differential expressions of synaptogenic markers between primary cultured cortical and hippocampal neurons
    • Kim, Y. G. and Lee, Y. I. (2012). Differential expressions of synaptogenic markers between primary cultured cortical and hippocampal neurons. Exp Neurobiol 21, 61-67. doi:10.5607/ en.2012.21.2.61
    • (2012) Exp Neurobiol , vol.21 , pp. 61-67
    • Kim, Y. G.1    Lee, Y. I.2
  • 157
    • 76749103795 scopus 로고    scopus 로고
    • MDMA (ecstasy) decreases the number of neurons and stem cells in embryonic cortical cultures
    • Kindlundh-Hogberg, A. M., Pickering, C., Wicher, G. et al. (2010). MDMA (ecstasy) decreases the number of neurons and stem cells in embryonic cortical cultures. Cell Mol Neurobiol 30, 13-21. doi:10.1007/s10571-009-9426-y
    • (2010) Cell Mol Neurobiol , vol.30 , pp. 13-21
    • Kindlundh-Hogberg, A. M.1    Pickering, C.2    Wicher, G.3
  • 158
    • 84994204383 scopus 로고    scopus 로고
    • Conversion of non-proliferating astrocytes into neurogenic neural stem cells: Control by FGF2 and IFN-gamma
    • Kleiderman, S., Gutbier, S., Tufekci, K. U. et al. (2016a). Conversion of non-proliferating astrocytes into neurogenic neural stem cells: Control by FGF2 and IFN-gamma. Stem Cells 34, 2861-2874. doi:10.1002/stem.2483
    • (2016) Stem Cells , vol.34 , pp. 2861-2874
    • Kleiderman, S.1    Gutbier, S.2    Tufekci, K. U.3
  • 159
    • 84952683446 scopus 로고    scopus 로고
    • Functional and phenotypic differences of pure populations of stem cell-derived astrocytes and neuronal precursor cells
    • Kleiderman, S., Sá, J. V., Teixeira, A. P. et al. (2016b). Functional and phenotypic differences of pure populations of stem cell-derived astrocytes and neuronal precursor cells. Glia 64, 695-715. doi:10.1002/glia.22954
    • (2016) Glia , vol.64 , pp. 695-715
    • Kleiderman, S.1    Sá, J. V.2    Teixeira, A. P.3
  • 160
    • 84876922933 scopus 로고    scopus 로고
    • A computational model predicting disruption of blood vessel development
    • Kleinstreuer, N., Dix, D., Rountree, M. et al. (2013). A computational model predicting disruption of blood vessel development. PLoS Comput Biol 9, e1002996. doi:10.1371/journal. pcbi.1002996
    • (2013) PLoS Comput Biol , vol.9 , pp. e1002996
    • Kleinstreuer, N.1    Dix, D.2    Rountree, M.3
  • 161
    • 81055145729 scopus 로고    scopus 로고
    • Brain plasticity and behaviour in the developing brain
    • Kolb, B. and Gibb, R. (2011). Brain plasticity and behaviour in the developing brain. J Can Acad Child Adolesc Psychiatry 20, 265-276.
    • (2011) J Can Acad Child Adolesc Psychiatry , vol.20 , pp. 265-276
    • Kolb, B.1    Gibb, R.2
  • 162
    • 33750375788 scopus 로고    scopus 로고
    • Patterns of neural stem and progenitor cell division may underlie evolutionary cortical expansion
    • Kriegstein, A., Noctor S. and Martinez-Cerdeno, V. (2006). Patterns of neural stem and progenitor cell division may underlie evolutionary cortical expansion. Nat Rev Neurosci 7, 883-890. doi:10.1038/nrn2008
    • (2006) Nat Rev Neurosci , vol.7 , pp. 883-890
    • Kriegstein, A.1    Noctor, S.2    Martinez-Cerdeno, V.3
  • 163
    • 84862693569 scopus 로고    scopus 로고
    • Systemic immune challenges trigger and drive Alzheimer-like neuropathology in mice
    • Krstic, D., Madhusudan, A., Doehner, J. et al. (2012). Systemic immune challenges trigger and drive Alzheimer-like neuropathology in mice. J Neuroinflammation 9, 151. doi: 10.1186/1742-2094-9-151
    • (2012) J Neuroinflammation , vol.9 , pp. 151
    • Krstic, D.1    Madhusudan, A.2    Doehner, J.3
  • 164
    • 84871922036 scopus 로고    scopus 로고
    • Deciphering the mechanism underlying late-onset Alzheimer disease
    • Krstic, D. and Knuesel, I. (2013). Deciphering the mechanism underlying late-onset Alzheimer disease. Nat Rev Neurol 9, 25-34. doi:10.1038/nrneurol.2012.236
    • (2013) Nat Rev Neurol , vol.9 , pp. 25-34
    • Krstic, D.1    Knuesel, I.2
  • 165
    • 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
  • 167
    • 83055180571 scopus 로고    scopus 로고
    • GFAP-independent inflammatory competence and trophic functions of astrocytes generated from murine embryonic stem cells
    • Kuegler, P. B., Baumann, B. A., Zimmer, B. et al. (2012). GFAP-independent inflammatory competence and trophic functions of astrocytes generated from murine embryonic stem cells. Glia 60, 218-228. doi:10.1002/glia.21257
    • (2012) Glia , vol.60 , pp. 218-228
    • Kuegler, P. B.1    Baumann, B. A.2    Zimmer, B.3
  • 168
    • 84884414984 scopus 로고    scopus 로고
    • Cerebral organoids model human brain development and microcephaly
    • Lancaster, M. A., Renner, M., Martin, C. A. et al. (2013). Cerebral organoids model human brain development and microcephaly. Nature 501, 373-379. doi:10.1038/nature12517
    • (2013) Nature , vol.501 , pp. 373-379
    • Lancaster, M. A.1    Renner, M.2    Martin, C. A.3
  • 169
    • 84887606653 scopus 로고    scopus 로고
    • A human pluripotent carcinoma stem cell-based model for in vitro developmental neurotoxicity testing: Effects of methylmercury, lead and aluminum evaluated by gene expression studies
    • Laurenza, I., Pallocca, G., Mennecozzi, M. et al. (2013). A human pluripotent carcinoma stem cell-based model for in vitro developmental neurotoxicity testing: Effects of methylmercury, lead and aluminum evaluated by gene expression studies. Int J Dev Neurosci 31, 679-691. doi:10.1016/j. ijdevneu.2013.03.002
    • (2013) Int J Dev Neurosci , vol.31 , pp. 679-691
    • Laurenza, I.1    Pallocca, G.2    Mennecozzi, M.3
  • 170
    • 84988664616 scopus 로고    scopus 로고
    • CYP3A5 mediates effects of cocaine on human neocorticogenesis: Studies using an in vitro 3D self-organized hPSC model with a single cortex-like unit
    • Lee, C. T., Chen, J., Kindberg, A. A. et al. (2017). CYP3A5 mediates effects of cocaine on human neocorticogenesis: Studies using an in vitro 3D self-organized hPSC model with a single cortex-like unit. Neuropsychopharmacology 42, 774-784. doi:/10.1038/npp.2016.156
    • (2017) Neuropsychopharmacology , vol.42 , pp. 774-784
    • Lee, C. T.1    Chen, J.2    Kindberg, A. A.3
  • 171
    • 70349301819 scopus 로고    scopus 로고
    • Modelling pathogenesis and treatment of familial dysautonomia using patient-specific iPSCs
    • Lee, G., Papapetrou, E. P., Kim, H. et al. (2009). Modelling pathogenesis and treatment of familial dysautonomia using patient-specific iPSCs. Nature 461, 402-406. doi:10.1038/ nature08320
    • (2009) Nature , vol.461 , pp. 402-406
    • Lee, G.1    Papapetrou, E. P.2    Kim, H.3
  • 172
    • 84905923814 scopus 로고    scopus 로고
    • Zebrafish as a model for investigating developmental lead (Pb) neurotoxicity as a risk factor in adult neurodegenerative disease: A mini-review
    • Lee, J. and Freeman J. L. (2014). Zebrafish as a model for investigating developmental lead (Pb) neurotoxicity as a risk factor in adult neurodegenerative disease: A mini-review. Neurotoxicology 43, 57-64. doi:/10.1016/j.neuro.2014.03.008
    • (2014) Neurotoxicology , vol.43 , pp. 57-64
    • Lee, J.1    Freeman, J. L.2
  • 173
    • 84945437300 scopus 로고    scopus 로고
    • Comparability of behavioural assays using zebrafish larvae to assess neurotoxicity
    • Legradi, J., el Abdellaoui, N., van Pomeren, M. and Legler, J. (2015). Comparability of behavioural assays using zebrafish larvae to assess neurotoxicity. Environ Sci Pollut Res Int 22, 16277-16289. doi:10.1007/s11356-014-3805-8
    • (2015) Environ Sci Pollut Res Int , vol.22 , pp. 16277-16289
    • Legradi, J.1    el Abdellaoui, N.2    van Pomeren, M.3    Legler, J.4
  • 174
    • 17044431495 scopus 로고    scopus 로고
    • In vitro and other alternative approaches to developmental neurotoxicity testing (DNT)
    • Lein, P., Silbergeld, E., Locke, P. and Goldberg, A. M. (2005). In vitro and other alternative approaches to developmental neurotoxicity testing (DNT). Environ Toxicol Pharmacol 19, 735-744. doi:10.1016/j.etap.2004.12.035
    • (2005) Environ Toxicol Pharmacol , vol.19 , pp. 735-744
    • Lein, P.1    Silbergeld, E.2    Locke, P.3    Goldberg, A. M.4
  • 175
    • 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
  • 176
    • 58949087263 scopus 로고    scopus 로고
    • The biological and ethical basis of the use of embryonic stem cells for in vitro tests and cell therapy
    • Leist, M., Bremer, S., Brundin, P. et al. (2008b). The biological and ethical basis of the use of embryonic stem cells for in vitro tests and cell therapy. ALTEX 25, 163-190. doi:10.14573/ altex.2008.3.163
    • (2008) ALTEX , vol.25 , pp. 163-190
    • Leist, M.1    Bremer, S.2    Brundin, P.3
  • 177
    • 79955432219 scopus 로고    scopus 로고
    • Food for thought on considerations and guidelines for basic test method descriptions in toxicology
    • Leist, M., Efremova, L. and Karreman, C. (2010). Food for thought on 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
  • 178
    • 84864593532 scopus 로고    scopus 로고
    • Validation and quality control of replacement alternatives – Current status and future challenges
    • Leist, M., Hasiwa, N., Daneshian, M. et al. (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
  • 179
    • 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
  • 180
    • 85031799653 scopus 로고    scopus 로고
    • Adverse outcome pathways: Opportunities, limitations and open questions
    • Leist, M., Ghallab, A., Graepel, R. et al. (2017). Adverse outcome pathways: Opportunities, limitations and open questions. Arch Toxicol 91, 3477-3505 doi:10.1007/s00204-017-2045-3
    • (2017) Arch Toxicol , vol.91 , pp. 3477-3505
    • Leist, M.1    Ghallab, A.2    Graepel, R.3
  • 181
    • 84892410589 scopus 로고    scopus 로고
    • The environmental neurotoxicant PCB 95 promotes synaptogenesis via ryanodine receptor-dependent miR132 upregulation
    • Lesiak, A., Zhu, M., Chen, H. et al. (2014). The environmental neurotoxicant PCB 95 promotes synaptogenesis via ryanodine receptor-dependent miR132 upregulation. J Neurosci 34, 717-725. doi:10.1523/JNEUROSCI.2884-13.2014
    • (2014) J Neurosci , vol.34 , pp. 717-725
    • Lesiak, A.1    Zhu, M.2    Chen, H.3
  • 182
    • 84971646057 scopus 로고    scopus 로고
    • Computational modeling and simulation of genital tubercle development
    • Leung, M. C. K., Hutson, M. S., Seifert, A. W. et al. (2016). Computational modeling and simulation of genital tubercle development. Reprod Toxicol 64, 151-161. doi:10.1016/j.reprotox.2016.05.005
    • (2016) Reprod Toxicol , vol.64 , pp. 151-161
    • Leung, M. C. K.1    Hutson, M. S.2    Seifert, A. W.3
  • 183
    • 84933673107 scopus 로고    scopus 로고
    • Conical expansion of the outer subventricular zone and the role of ceocortical folding in evolution and development
    • Lewitus, E., Kelave, I. and Huttner, W. B. (2013). Conical expansion of the outer subventricular zone and the role of ceocortical folding in evolution and development. Front Hum Neurosci 7, 424. doi:10.3389/fnhum.2013.00424
    • (2013) Front Hum Neurosci , vol.7 , pp. 424
    • Lewitus, E.1    Kelave, I.2    Huttner, W. B.3
  • 184
    • 84988556744 scopus 로고    scopus 로고
    • Developmental exposure to paraquat and maneb can impair cognition, learning and memory in Sprague-Dawley rats
    • Li, B., He, X., Sun, Y. and Li, B. (2016). Developmental exposure to paraquat and maneb can impair cognition, learning and memory in Sprague-Dawley rats. Mol Biosyst 12, 3088-3097. doi:10.1039/C6MB00284F
    • (2016) Mol Biosyst , vol.12 , pp. 3088-3097
    • Li, B.1    He, X.2    Sun, Y.3    Li, B.4
  • 185
    • 0036279763 scopus 로고    scopus 로고
    • PTEN in neural precursor cells: Regulation of migration, apoptosis, and proliferation
    • Li, L., Liu, F., Salmonsen, R. A. et al. (2002). PTEN in neural precursor cells: Regulation of migration, apoptosis, and proliferation. Mol Cell Neurosci 20, 21-29. doi:10.1006/ mcne.2002.1115
    • (2002) Mol Cell Neurosci , vol.20 , pp. 21-29
    • Li, L.1    Liu, F.2    Salmonsen, R. A.3
  • 186
    • 85016155890 scopus 로고    scopus 로고
    • Neurotoxic reactive astrocytes are induced by activated microglia
    • Liddelow, S. A., Guttenplan, K. A., Clarke, L. E. et al. (2017). Neurotoxic reactive astrocytes are induced by activated microglia. Nature 541, 481-487. doi:10.1038/nature21029
    • (2017) Nature , vol.541 , pp. 481-487
    • Liddelow, S. A.1    Guttenplan, K. A.2    Clarke, L. E.3
  • 187
    • 84908232014 scopus 로고    scopus 로고
    • The neurology of mTOR
    • Lipton, J. O. and Sahin, M. (2014). The neurology of mTOR. Neuron 84, 275-291. doi:10.1016/j.neuron.2014.09.034
    • (2014) Neuron , vol.84 , pp. 275-291
    • Lipton, J. O.1    Sahin, M.2
  • 188
    • 0037101970 scopus 로고    scopus 로고
    • Function and regulation of CREB family transcription factors in the nervous system
    • Lonze, B. E. and Ginty, D. D. (2002). Function and regulation of CREB family transcription factors in the nervous system. Neuron 35, 605-623. doi:10.1016/S0896-6273(02)00828-0
    • (2002) Neuron , vol.35 , pp. 605-623
    • Lonze, B. E.1    Ginty, D. D.2
  • 189
    • 84916637399 scopus 로고    scopus 로고
    • Mutations of the thyroid hormone transporter MCT8 cause prenatal brain damage and persistent hypomyelination
    • López-Espíndola, D., Morales-Bastos, C., Grijota-Martínez, C. et al. (2014). Mutations of the thyroid hormone transporter MCT8 cause prenatal brain damage and persistent hypomyelination. J Clin Endocrinol Metab 99, E2799-2804. doi:10.1210/jc.2014-2162
    • (2014) J Clin Endocrinol Metab , vol.99 , pp. E2799-E2804
    • López-Espíndola, D.1    Morales-Bastos, C.2    Grijota-Martínez, C.3
  • 190
    • 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
  • 191
    • 39849083186 scopus 로고    scopus 로고
    • BDNF: A key regulator for protein synthesis dependent LTP and long-term memory?
    • Lu, Y., Christian, K. and Lu, B. (2008). BDNF: A key regulator for protein synthesis dependent LTP and long-term memory? Neurobiol Learn Mem 89, 312-323. doi:10.1016/j. nlm.2007.08.018
    • (2008) Neurobiol Learn Mem , vol.89 , pp. 312-323
    • Lu, Y.1    Christian, K.2    Lu, B.3
  • 192
    • 33750627921 scopus 로고    scopus 로고
    • The dynamics of the LPS triggered inflammatory response of murine microglia under different culture and in vivo conditions
    • Lund, S., Porzgen, P., Mortensen, A. L. et al. (2006). The dynamics of the LPS triggered inflammatory response of murine microglia under different culture and in vivo conditions. J Neuroimmunol 180, 71-87. doi:10.1016/j.jneuroim.2006.07.007
    • (2006) J Neuroimmunol , vol.180 , pp. 71-87
    • Lund, S.1    Porzgen, P.2    Mortensen, A. L.3
  • 193
    • 84988410239 scopus 로고    scopus 로고
    • In vitro and ex vivo models of multiple sclerosis
    • Madill, M., Fitzgerald, D., O’Connell, K. E. et al. (2016). In vitro and ex vivo models of multiple sclerosis. Drug Discov Today 21, 1504-1511. doi:10.1016/j.drudis.2016.05.018
    • (2016) Drug Discov Today , vol.21 , pp. 1504-1511
    • Madill, M.1    Fitzgerald, D.2    O’Connell, K. E.3
  • 194
    • 85018509261 scopus 로고    scopus 로고
    • AKT/PKB signaling: Navigating the network
    • Manning, B. D. and Toker, A. (2017). AKT/PKB signaling: Navigating the network. Cell 169, 381-405. doi:10.1016/j. cell.2017.04.001
    • (2017) Cell , vol.169 , pp. 381-405
    • Manning, B. D.1    Toker, A.2
  • 195
    • 78650296101 scopus 로고    scopus 로고
    • beta-adrenoceptor activation depresses brain inflammation and is neuroprotective in lipopolysaccharide-induced sensitization to oxygen-glucose deprivation in organotypic hippocampal slices
    • Markus, T., Hansson, S. R., Cronberg, T. et al. (2010). beta-adrenoceptor activation depresses brain inflammation and is neuroprotective in lipopolysaccharide-induced sensitization to oxygen-glucose deprivation in organotypic hippocampal slices. J Neuroinflammation 7, 94. doi:10.1186/1742-2094-7-94
    • (2010) J Neuroinflammation , vol.7 , pp. 94
    • Markus, T.1    Hansson, S. R.2    Cronberg, T.3
  • 196
    • 84979255712 scopus 로고    scopus 로고
    • Biology-inspired microphysiological system approaches to solve the prediction dilemma of substance testing using animals
    • Marx, U., Andersson, T. B., Bahinski, A. et al. (2016). Biology-inspired microphysiological system approaches to solve the prediction dilemma of substance testing using animals. 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
  • 197
    • 69849088915 scopus 로고    scopus 로고
    • A review of the implementation of the embryonic stem cell test (EST). The report and recommendations of an ECVAM/ReProTect workshop
    • Marx-Stoelting, P., Adriaens, E., Ahr, H. J. et al. (2009). A review of the implementation of the embryonic stem cell test (EST). The report and recommendations of an ECVAM/ReProTect workshop. Altern Lab Anim 37, 313-328.
    • (2009) Altern Lab Anim , vol.37 , pp. 313-328
    • Marx-Stoelting, P.1    Adriaens, E.2    Ahr, H. J.3
  • 198
    • 56649124807 scopus 로고    scopus 로고
    • Glutamate and neurotrophic factors in neuronal plasticity and disease
    • Mattson, M. P. (2008). Glutamate and neurotrophic factors in neuronal plasticity and disease. Ann N Y Acad Sci 1144, 97-112. doi:10.1196/annals.1418.005
    • (2008) Ann N Y Acad Sci , vol.1144 , pp. 97-112
    • Mattson, M. P.1
  • 199
    • 80053049161 scopus 로고    scopus 로고
    • Cocaine alters BDNF expression and neuronal migration in the embryonic mouse forebrain
    • McCarthy, D. M., Zhang, X., Darnell, S. B. et al. (2011). Cocaine alters BDNF expression and neuronal migration in the embryonic mouse forebrain. J Neurosci 31, 13400-13411. doi:10.1523/JNEUROSCI.2944-11.2011
    • (2011) J Neurosci , vol.31 , pp. 13400-13411
    • McCarthy, D. M.1    Zhang, X.2    Darnell, S. B.3
  • 200
    • 38349049457 scopus 로고    scopus 로고
    • Estradiol and the developing brain
    • McCarthy, M. M. (2008). Estradiol and the developing brain. Physiol Rev 88, 91-124. doi:10.1152/physrev.00010.2007
    • (2008) Physiol Rev , vol.88 , pp. 91-124
    • McCarthy, M. M.1
  • 201
    • 84930082946 scopus 로고    scopus 로고
    • Gaining acceptance for the use of in vitro toxicity assays and QIVIVE in regulatory risk assessment
    • Meek, M. E. and Lipscomb, J. C. (2015). Gaining acceptance for the use of in vitro toxicity assays and QIVIVE in regulatory risk assessment. Toxicology 332, 112-123. doi:10.1016/j. tox.2015.01.010
    • (2015) Toxicology , vol.332 , pp. 112-123
    • Meek, M. E.1    Lipscomb, J. C.2
  • 202
    • 18244395545 scopus 로고    scopus 로고
    • Craniofacial and axial skeletal defects induced by the fungicide triadimefon in the mouse
    • Menegola, E., Broccia, M. L., Di Renzo, F. et al. (2005). Craniofacial and axial skeletal defects induced by the fungicide triadimefon in the mouse. Birth Defects Res B Dev Reprod Toxicol 74, 185-195. doi:10.1002/bdrb.20035
    • (2005) Birth Defects Res B Dev Reprod Toxicol , vol.74 , pp. 185-195
    • Menegola, E.1    Broccia, M. L.2    Di Renzo, F.3
  • 203
    • 84900303453 scopus 로고    scopus 로고
    • Transcriptional landscape of the prenatal human brain
    • Miller, J. A., Ding, S. L., Sunkin, S. M. et al. (2014). Transcriptional landscape of the prenatal human brain. Nature 508, 199-206. doi:10.1038/nature13185
    • (2014) Nature , vol.508 , pp. 199-206
    • Miller, J. A.1    Ding, S. L.2    Sunkin, S. M.3
  • 204
    • 84930869090 scopus 로고    scopus 로고
    • Astrocyte development: A guide for the perplexed
    • Molofsky, A. V. and Deneend, B. (2015). Astrocyte development: A guide for the perplexed. Glia 63, 1320-1329. doi:10.1002/ glia.22836
    • (2015) Glia , vol.63 , pp. 1320-1329
    • Molofsky, A. V.1    Deneend, B.2
  • 205
    • 0027483917 scopus 로고
    • Evaluation of the toxicity of different metal compounds in the developing brain using aggregating cell cultures as a model
    • Monnet-Tschudi, F., Zurich, M. G. and Honegger, P. (1993). Evaluation of the toxicity of different metal compounds in the developing brain using aggregating cell cultures as a model. Toxicol In Vitro 7, 335-339. doi:10.1016/0887-2333(93)90024-Y
    • (1993) Toxicol In Vitro , vol.7 , pp. 335-339
    • Monnet-Tschudi, F.1    Zurich, M. G.2    Honegger, P.3
  • 206
    • 0030602015 scopus 로고    scopus 로고
    • Comparison of the developmental effects of two mercury compounds on glial cells and neurons in aggregate cultures of rat telencephalon
    • Monnet-Tschudi, F., Zurich, M. G. and Honegger, P. (1996). Comparison of the developmental effects of two mercury compounds on glial cells and neurons in aggregate cultures of rat telencephalon. Brain Res 741, 52-59. doi:10.1016/S0006-8993(96)00895-5
    • (1996) Brain Res , vol.741 , pp. 52-59
    • Monnet-Tschudi, F.1    Zurich, M. G.2    Honegger, P.3
  • 207
    • 0032972025 scopus 로고    scopus 로고
    • The naturally occurring food mycotoxin fumonisin B1 impairs myelin formation in aggregating brain cell culture
    • Monnet-Tschudi, F., Zurich, M. G., Sorg, O. et al. (1999). The naturally occurring food mycotoxin fumonisin B1 impairs myelin formation in aggregating brain cell culture. Neurotoxicology 20, 41-48.
    • (1999) Neurotoxicology , vol.20 , pp. 41-48
    • Monnet-Tschudi, F.1    Zurich, M. G.2    Sorg, O.3
  • 208
    • 0034659286 scopus 로고    scopus 로고
    • Maturation-dependent effects of chlorpyrifos and parathion and their oxygen analogs on acetylcholinesterase and neuronal and glial markers in aggregating brain cell cultures
    • Monnet-Tschudi, F., Zurich, M. G., Schilter, B. et al. (2000). Maturation-dependent effects of chlorpyrifos and parathion and their oxygen analogs on acetylcholinesterase and neuronal and glial markers in aggregating brain cell cultures. Toxicol Appl Pharmacol 165, 175-183. doi:10.1006/taap.2000.8934
    • (2000) Toxicol Appl Pharmacol , vol.165 , pp. 175-183
    • Monnet-Tschudi, F.1    Zurich, M. G.2    Schilter, B.3
  • 209
    • 3242666768 scopus 로고    scopus 로고
    • Essential role of brain-derived neurotrophic factor in adult hippocampal function
    • Monteggia, L. M., Barrot, M., Powell, C. M. et al. (2004). Essential role of brain-derived neurotrophic factor in adult hippocampal function. Proc Natl Acad Sci U S A 101, 10827-10832. doi:10.1073/pnas.0402141101
    • (2004) Proc Natl Acad Sci U S A , vol.101 , pp. 10827-10832
    • Monteggia, L. M.1    Barrot, M.2    Powell, C. M.3
  • 210
    • 34247893688 scopus 로고    scopus 로고
    • ERK-dependent and -independent pathways trigger human neural progenitor cell migration
    • Moors, M., Cline, J. E., Abel, J. and Fritsche, E. (2007). ERK-dependent and -independent pathways trigger human neural progenitor cell migration. Toxicol Appl Pharmacol 221, 57-67. doi:10.1016/j.taap.2007.02.018
    • (2007) Toxicol Appl Pharmacol , vol.221 , pp. 57-67
    • Moors, M.1    Cline, J. E.2    Abel, J.3    Fritsche, E.4
  • 211
    • 67650891858 scopus 로고    scopus 로고
    • Human neurospheres as three-dimensional cellular systems for developmental neurotoxicity testing
    • Moors, M., Rockel, T. D., Abel, J. et al. (2009). Human neurospheres as three-dimensional cellular systems for developmental neurotoxicity testing. Environ Health Perspect 117, 1131-1138. doi:10.1289/ehp.0800207
    • (2009) Environ Health Perspect , vol.117 , pp. 1131-1138
    • Moors, M.1    Rockel, T. D.2    Abel, J.3
  • 212
    • 75049085393 scopus 로고    scopus 로고
    • Interleukin-7 (IL-7) and IL-7 splice variants affect differentiation of human neural progenitor cells
    • Moors, M., Vudattu, N. K., Abel, J. et al. (2010). Interleukin-7 (IL-7) and IL-7 splice variants affect differentiation of human neural progenitor cells. Genes Immun 11, 11-20. doi:10.1038/ gene.2009.77
    • (2010) Genes Immun , vol.11 , pp. 11-20
    • Moors, M.1    Vudattu, N. K.2    Abel, J.3
  • 213
    • 84856091200 scopus 로고    scopus 로고
    • Dickkopf 1 mediates glucocorticoid-induced changes in human neural progenitor cell proliferation and differentiation
    • Moors, M., Bose, R., Johansson-Hague, K. et al. (2012). Dickkopf 1 mediates glucocorticoid-induced changes in human neural progenitor cell proliferation and differentiation. Toxicol Sci 125, 488-495. doi:10.1093/toxsci/kfr304
    • (2012) Toxicol Sci , vol.125 , pp. 488-495
    • Moors, M.1    Bose, R.2    Johansson-Hague, K.3
  • 214
    • 74949099848 scopus 로고    scopus 로고
    • 2+/calmodulin-dependent protein kinase IV pathway
    • 2+/calmodulin-dependent protein kinase IV pathway. Endocrinology 151, 810-820. doi:10.1210/en.2009-0958
    • (2010) Endocrinology , vol.151 , pp. 810-820
    • Morte, B.1    Díez, D.2    Ausó, E.3
  • 216
    • 77950020507 scopus 로고    scopus 로고
    • Neuronal models for evaluation of proliferation in vitro using high content screening
    • Mundy, W. R., Radio, N. M. and Freudenrich, T. M. (2010). Neuronal models for evaluation of proliferation in vitro using high content screening. Toxicology 270, 121-130. doi:10.1016/j. tox.2010.02.004
    • (2010) Toxicology , vol.270 , pp. 121-130
    • Mundy, W. R.1    Radio, N. M.2    Freudenrich, T. M.3
  • 217
    • 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
  • 218
    • 23944445784 scopus 로고    scopus 로고
    • Activity-dependent modulation of the BDNF receptor TrkB: Mechanisms and implications
    • Nagappan, G. and Lu, B. (2005). Activity-dependent modulation of the BDNF receptor TrkB: Mechanisms and implications. Trends Neurosci 28, 464-471. doi:10.1016/j.tins.2005.07.003
    • (2005) Trends Neurosci , vol.28 , pp. 464-471
    • Nagappan, G.1    Lu, B.2
  • 219
    • 84857500997 scopus 로고    scopus 로고
    • Human embryonic stem cell model of ethanol-mediated early developmental toxicity
    • Nash, R., Krishnamoorthy, M., Jenkins, A. and Csete, M. (2012). Human embryonic stem cell model of ethanol-mediated early developmental toxicity. Exp Neurol 234, 127-135. doi:10.1016/j.expneurol.2011.12.022
    • (2012) Exp Neurol , vol.234 , pp. 127-135
    • Nash, R.1    Krishnamoorthy, M.2    Jenkins, A.3    Csete, M.4
  • 220
    • 78650993109 scopus 로고    scopus 로고
    • 2+): Effect on synaptic function
    • 2+): Effect on synaptic function. Mol Neurobiol 42, 151-160. doi:10.1007/s00210-008-0344-1
    • (2010) Mol Neurobiol , vol.42 , pp. 151-160
    • Neal, A. P.1    Guillarte, R. T.2
  • 221
    • 84941578670 scopus 로고    scopus 로고
    • Molecular regulation of synaptogenesis during associative learning and memory
    • Nelson, T. J. and Alkon, D. L. (2015). Molecular regulation of synaptogenesis during associative learning and memory. Brain Res 1621, 239-251 doi:10.1016/j.brainres.2014.11.054
    • (2015) Brain Res , vol.1621 , pp. 239-251
    • Nelson, T. J.1    Alkon, D. L.2
  • 222
    • 84919740393 scopus 로고    scopus 로고
    • Glucocorticoids promote neural progenitor cell proliferation derived from human induced pluripotent stem cells
    • Ninomiya, E., Hattori, T., Toyoda, M. et al. (2014). Glucocorticoids promote neural progenitor cell proliferation derived from human induced pluripotent stem cells. Springerplus 3, 527. doi:10.1186/2193-1801-3-527
    • (2014) Springerplus , vol.3 , pp. 527
    • Ninomiya, E.1    Hattori, T.2    Toyoda, M.3
  • 223
    • 84920983678 scopus 로고    scopus 로고
    • Zebrafish as a systems toxicology model for developmental neurotoxicity testing
    • Nishimura, Y., Murakami, S., Ashikawa, Y. et al. (2015). Zebrafish as a systems toxicology model for developmental neurotoxicity testing. Congenit Anom (Kyoto) 55, 1-16. doi:10.1111/ cga.12079
    • (2015) Congenit Anom (Kyoto) , vol.55 , pp. 1-16
    • Nishimura, Y.1    Murakami, S.2    Ashikawa, Y.3
  • 224
    • 84929665664 scopus 로고    scopus 로고
    • Advanced morphological – Behavioral test platform reveals neurodevelopmental defects in embryonic zebrafish exposed to comprehensive suite of halogenated and organophosphate flame retardants
    • Noyes, P. D., Haggard, D. E., Gonnerman, G. D. and Tanguay, R. L. (2015). Advanced morphological – Behavioral test platform reveals neurodevelopmental defects in embryonic zebrafish exposed to comprehensive suite of halogenated and organophosphate flame retardants. Toxicol Sci 145, 177-195. doi:10.1093/toxsci/kfv044
    • (2015) Toxicol Sci , vol.145 , pp. 177-195
    • Noyes, P. D.1    Haggard, D. E.2    Gonnerman, G. D.3    Tanguay, R. L.4
  • 225
    • 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. (2017a). 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
  • 226
    • 85018724518 scopus 로고    scopus 로고
    • Combination of multiple neural crest migration assay to identify environmental toxicants from a proof-of-concept chemical library
    • Nyffeler, J., Dolde, X., Krebs, A. et al. (2017b). Combination of multiple neural crest migration assay 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
  • 227
    • 0346665563 scopus 로고    scopus 로고
    • Hormone-dependent repression of the E2F-1 gene by thyroid hormone receptors
    • Nygard, M., Wahlstrom, G. M., Gustafsson, M. V. et al. (2003). Hormone-dependent repression of the E2F-1 gene by thyroid hormone receptors. Mol Endocrinol 17, 79-92. doi:10.1210/ me.2002-0107
    • (2003) Mol Endocrinol , vol.17 , pp. 79-92
    • Nygard, M.1    Wahlstrom, G. M.2    Gustafsson, M. V.3
  • 231
    • 84886162191 scopus 로고    scopus 로고
    • ENV/JM/MONO(2014)35. Series on Testing and Assessment No. 211, OECD Publishing, Paris
    • OECD (2014b). Guidance Document for Describing Non-Guideline In Vitro Test Methods. ENV/JM/MONO(2014)35. Series on Testing and Assessment No. 211, OECD Publishing, Paris. doi:10.1787/20777876
    • (2014) Guidance Document for Describing Non-Guideline In Vitro Test Methods
  • 236
    • 84885133606 scopus 로고    scopus 로고
    • Retinoid machinery in distinct neural stem cell populations with different retinoid responsiveness
    • Orsolits, B., Borsy, A., Madarasz, E. et al. (2013). Retinoid machinery in distinct neural stem cell populations with different retinoid responsiveness. Stem Cells Dev 22, 2777-2793. doi:10.1089/scd.2012.0422
    • (2013) Stem Cells Dev , vol.22 , pp. 2777-2793
    • Orsolits, B.1    Borsy, A.2    Madarasz, E.3
  • 237
    • 84855933284 scopus 로고    scopus 로고
    • Assessing locomotor activity in larval zebrafish: Influence of extrinsic and intrinsic variables
    • Padilla, S., Hunter, D. L., Padnos, B. et al. (2011). Assessing locomotor activity in larval zebrafish: Influence of extrinsic and intrinsic variables. Neurotoxicol Teratol 33, 624-630. doi:10.1016/j.ntt.2011.08.005
    • (2011) Neurotoxicol Teratol , vol.33 , pp. 624-630
    • Padilla, S.1    Hunter, D. L.2    Padnos, B.3
  • 238
    • 79952718925 scopus 로고    scopus 로고
    • Human embryonic stem cell proliferation and differentiation as parameters to evaluate developmental toxicity
    • Pal, R., Mamidi, M. K., Das, A. K. and Bhonde, R. (2011). Human embryonic stem cell proliferation and differentiation as parameters to evaluate developmental toxicity. J Cell Physiol 226, 1583-1595. doi:10.1002/jcp.22484
    • (2011) J Cell Physiol , vol.226 , pp. 1583-1595
    • Pal, R.1    Mamidi, M. K.2    Das, A. K.3    Bhonde, R.4
  • 239
    • 84883306186 scopus 로고    scopus 로고
    • miRNA expression profiling in a human stem cell-based model as a tool for developmental neurotoxicity testing
    • Pallocca, G., Fabbri, M., Sacco, M. G. et al. (2013). miRNA expression profiling in a human stem cell-based model as a tool for developmental neurotoxicity testing. Cell Biol Toxicol 29, 239-257, doi:10.1007/s10565-013-9250-5
    • (2013) Cell Biol Toxicol , vol.29 , pp. 239-257
    • Pallocca, G.1    Fabbri, M.2    Sacco, M. G.3
  • 240
    • 84955382107 scopus 로고    scopus 로고
    • Identification of transcriptome signatures and biomarkers specific for potential developmental toxicants inhibiting human neural crest cell migration
    • Pallocca, G., Grinberg, M., Henry, M. et al. (2016). Identification of transcriptome signatures and biomarkers specific for potential developmental toxicants inhibiting human neural crest cell migration. Arch Toxicol 90, 159-180. doi:10.1007/ s00204-015-1658-7
    • (2016) Arch Toxicol , vol.90 , pp. 159-180
    • Pallocca, G.1    Grinberg, M.2    Henry, M.3
  • 241
    • 84864551960 scopus 로고    scopus 로고
    • Metabolic biomarkers of prenatal alcohol exposure in human embryonic stem cell-derived neural lineages
    • Palmer, J. A., Poenitzsch, A. M., Smith, S. M. et al. (2012). Metabolic biomarkers of prenatal alcohol exposure in human embryonic stem cell-derived neural lineages. Alcohol Clin Exp Res 36, 1314-1324. doi:10.1111/j.1530-0277.2011.01732.x
    • (2012) Alcohol Clin Exp Res , vol.36 , pp. 1314-1324
    • Palmer, J. A.1    Poenitzsch, A. M.2    Smith, S. M.3
  • 242
    • 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. (2017a). 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
    • (2017) ALTEX , vol.34 , pp. 362-376
    • Pamies, D.1    Barreras, P.2    Block, K.3
  • 243
    • 85012027021 scopus 로고    scopus 로고
    • Good cell culture practice for stem cells and stem-cell-derived models
    • Pamies, D., Bal-Price, A., Simeonov, A. et al. (2017b). Good cell culture practice for stem cells and stem-cell-derived models. ALTEX 34, 95-132. doi:10.14573/altex.1607121
    • (2017) ALTEX , vol.34 , pp. 95-132
    • Pamies, D.1    Bal-Price, A.2    Simeonov, A.3
  • 244
  • 245
    • 77955844066 scopus 로고    scopus 로고
    • The comparative neuroanatomy and neurochemistry of zebrafish CNS systems of relevance to human neuropsychiatric diseases
    • Panula, P., Chen, Y. C., Priyadarshini, M. et al. (2010). The comparative neuroanatomy and neurochemistry of zebrafish CNS systems of relevance to human neuropsychiatric diseases. Neurobiol Dis 40, 46-57. doi:10.1016/j.nbd.2010.05.010
    • (2010) Neurobiol Dis , vol.40 , pp. 46-57
    • Panula, P.1    Chen, Y. C.2    Priyadarshini, M.3
  • 246
    • 84871408179 scopus 로고    scopus 로고
    • Neurotrophin regulation of neural circuit development and function
    • Park, H. and Poo, M. M. (2013). Neurotrophin regulation of neural circuit development and function. Nat Rev Neurosci 14, 7-23. doi:10.1038/nrn3379
    • (2013) Nat Rev Neurosci , vol.14 , pp. 7-23
    • Park, H.1    Poo, M. M.2
  • 247
    • 84875513880 scopus 로고    scopus 로고
    • The role of zebrafish (Danio rerio) in dissecting the genetics and neural circuits of executive function
    • Parker, M. O., Brock, A. J., Walton, R. T. et al. (2013). The role of zebrafish (Danio rerio) in dissecting the genetics and neural circuits of executive function. Front Neural Circuits 7, 63. doi:10.3389/fncir.2013.00063
    • (2013) Front Neural Circuits , vol.7 , pp. 63
    • Parker, M. O.1    Brock, A. J.2    Walton, R. T.3
  • 248
    • 84908115946 scopus 로고    scopus 로고
    • The utility of zebrafish to study the mechanisms by which ethanol affects social behavior and anxiety during early brain development
    • Parker, M. O., Annan, L. V., Kanellopoulos, A. H. et al. (2014). The utility of zebrafish to study the mechanisms by which ethanol affects social behavior and anxiety during early brain development. Prog Neuropsychopharmacol Biol Psychiatry 55, 94-100, doi:10.1016/j.pnpbp.2014.03.011
    • (2014) Prog Neuropsychopharmacol Biol Psychiatry , vol.55 , pp. 94-100
    • Parker, M. O.1    Annan, L. V.2    Kanellopoulos, A. H.3
  • 249
    • 84904051395 scopus 로고    scopus 로고
    • Towards AOP application – Implementation of an integrated approach to testing and assessment (IATA) into a pipeline tool for skin sensitization
    • Patlewicz, G., Kuseva, C., Kesova, A. et al. (2014). Towards AOP application – Implementation of an integrated approach to testing and assessment (IATA) into a pipeline tool for skin sensitization. Regul Toxicol Pharmacol 69, 529-545. doi:10.1016/j.yrtph.2014.06.001
    • (2014) Regul Toxicol Pharmacol , vol.69 , pp. 529-545
    • Patlewicz, G.1    Kuseva, C.2    Kesova, A.3
  • 250
    • 85031421199 scopus 로고    scopus 로고
    • Evaluation and calibration of high-throughput predictions of chemical distribution to tissues
    • Pearce, R. G., Setzer, R. W., Davis, J. L. and Wambaugh, J. F. (2017). Evaluation and calibration of high-throughput predictions of chemical distribution to tissues. J Pharmacokinet Pharmacodyn 44, 549-565. doi:10.1007/s10928-017-9548-7
    • (2017) J Pharmacokinet Pharmacodyn , vol.44 , pp. 549-565
    • Pearce, R. G.1    Setzer, R. W.2    Davis, J. L.3    Wambaugh, J. F.4
  • 251
    • 84858619494 scopus 로고    scopus 로고
    • Enhanced neurogenesis in organotypic cultures of rat hippocampus after transient subfield-selective excitotoxic insult induced by domoic acid
    • Perez-Gomez, A. and Tasker, R. A. (2012). Enhanced neurogenesis in organotypic cultures of rat hippocampus after transient subfield-selective excitotoxic insult induced by domoic acid. Neuroscience 208, 97-108. doi:10.1016/j. neuroscience.2012.02.003
    • (2012) Neuroscience , vol.208 , pp. 97-108
    • Perez-Gomez, A.1    Tasker, R. A.2
  • 252
    • 84876341495 scopus 로고    scopus 로고
    • Evaluation of an alternative in vitro test battery for detecting reproductive toxicants
    • Piersma, A. H., Bosgra, S., van Duursen, M. B. et al. (2013). Evaluation of an alternative in vitro test battery for detecting reproductive toxicants. Reprod Toxicol 38, 53-64. doi:10.1016/j.reprotox.2013.03.002
    • (2013) Reprod Toxicol , vol.38 , pp. 53-64
    • Piersma, A. H.1    Bosgra, S.2    van Duursen, M. B.3
  • 253
    • 84910038158 scopus 로고    scopus 로고
    • A critical appraisal of the process of regulatory implementation of novel in vivo and in vitro methods for chemical hazard and risk assessment
    • Piersma, A. H., Ezendam, J., Luijten, M. et al. (2014). A critical appraisal of the process of regulatory implementation of novel in vivo and in vitro methods for chemical hazard and risk assessment. Crit Rev Toxicol 44, 876-894. doi:10.3109/104084 44.2014.940445
    • (2014) Crit Rev Toxicol , vol.44 , pp. 876-894
    • Piersma, A. H.1    Ezendam, J.2    Luijten, M.3
  • 254
    • 84908388684 scopus 로고    scopus 로고
    • Development of a pluripotent stem cell derived neuronal model to identify chemically induced pathway perturbations in relation to neurotoxicity: Effects of CREB pathway inhibition
    • Pistollato, F., Louisse, J., Scelfo, B. et al. (2014). Development of a pluripotent stem cell derived neuronal model to identify chemically induced pathway perturbations in relation to neurotoxicity: Effects of CREB pathway inhibition. Toxicol Appl Pharmacol 280, 378-388. doi:10.1016/j.taap.2014.08.007
    • (2014) Toxicol Appl Pharmacol , vol.280 , pp. 378-388
    • Pistollato, F.1    Louisse, J.2    Scelfo, B.3
  • 255
    • 85021244524 scopus 로고    scopus 로고
    • Protocol for the differentiation of human induced pluripotent stem cells into mixed cultures of neurons and glia for neurotoxicity testing
    • Pistollato, F., Canovas-Jorda, D., Zagoura, D. and Price, A. (2017). Protocol for the differentiation of human induced pluripotent stem cells into mixed cultures of neurons and glia for neurotoxicity testing. J Vis Exp 124, e55702. doi:10.3791/55702
    • (2017) J Vis Exp , vol.124 , pp. e55702
    • Pistollato, F.1    Canovas-Jorda, D.2    Zagoura, D.3    Price, A.4
  • 256
    • 84994750497 scopus 로고    scopus 로고
    • Advancing toxicology research using in vivo high throughput toxicology with small fish models
    • Planchart, A., Mattingly, C., Allen, D. et al. (2016). Advancing toxicology research using in vivo high throughput toxicology with small fish models. ALTEX 33, 435-452. doi:10.14573/ altex.1601281
    • (2016) ALTEX , vol.33 , pp. 435-452
    • Planchart, A.1    Mattingly, C.2    Allen, D.3
  • 257
    • 84892491604 scopus 로고    scopus 로고
    • A fluorescence micro-plate screen assay for the detection of neurite outgrowth and neurotoxicity using an antibody against betaIII-tubulin
    • Popova, D. and Jacobsson, S. O. (2014). A fluorescence micro-plate screen assay for the detection of neurite outgrowth and neurotoxicity using an antibody against betaIII-tubulin. Toxicol In Vitro 28, 411-418. doi:10.1016/j.tiv.2013.12.009
    • (2014) Toxicol In Vitro , vol.28 , pp. 411-418
    • Popova, D.1    Jacobsson, S. O.2
  • 258
    • 84875643749 scopus 로고    scopus 로고
    • EGF transactivation of Trk receptors regulates the migration of newborn cortical neurons
    • Puehringer, D., Orel, N., Lüningschrör, P. et al. (2013). EGF transactivation of Trk receptors regulates the migration of newborn cortical neurons. Nat Neurosci 16, 407-415. doi:10.1038/nn.3333
    • (2013) Nat Neurosci , vol.16 , pp. 407-415
    • Puehringer, D.1    Orel, N.2    Lüningschrör, P.3
  • 259
    • 0036158822 scopus 로고    scopus 로고
    • Chemotherapy-induced peripheral neuropathy
    • Quasthoff, S. and Hartung, H. P. (2002). Chemotherapy-induced peripheral neuropathy. J Neurol 249, 9-17. doi:10.1007/ PL00007853
    • (2002) J Neurol , vol.249 , pp. 9-17
    • Quasthoff, S.1    Hartung, H. P.2
  • 260
    • 84964619895 scopus 로고    scopus 로고
    • Brain-region-specific organoids using mini-bioreactors for modeling ZIKV exposure
    • Qian, X., Nguyen, H. N., Song, M. M. et al. (2016). Brain-region-specific organoids using mini-bioreactors for modeling ZIKV exposure. Cell 165, 1238-1254. doi:10.1016/j. cell.2016.04.032
    • (2016) Cell , vol.165 , pp. 1238-1254
    • Qian, X.1    Nguyen, H. N.2    Song, M. M.3
  • 261
    • 84977845297 scopus 로고    scopus 로고
    • Use of neural models of proliferation and neurite outgrowth to screen environmental chemicals in the ToxCast phase I library
    • Radio, N. M., Breier, J. M., Reif, D. M. et al. (2015). Use of neural models of proliferation and neurite outgrowth to screen environmental chemicals in the ToxCast phase I library. Appl In Vitro Toxicol 1, 131-139. doi:10.1089/aivt.2014.0009
    • (2015) Appl In Vitro Toxicol , vol.1 , pp. 131-139
    • Radio, N. M.1    Breier, J. M.2    Reif, D. M.3
  • 262
    • 77955514436 scopus 로고    scopus 로고
    • The use of developmental neurotoxicity data in pesticide risk assessments
    • Raffaele, K. C., Rowland, J., May, B. et al. (2010). The use of developmental neurotoxicity data in pesticide risk assessments. Neurotoxicol Teratol 32, 563-572. doi:10.1016/j. ntt.2010.04.053
    • (2010) Neurotoxicol Teratol , vol.32 , pp. 563-572
    • Raffaele, K. C.1    Rowland, J.2    May, B.3
  • 263
    • 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
  • 264
    • 78650284457 scopus 로고    scopus 로고
    • Endocrine profiling and prioritization of environmental chemicals using ToxCast data
    • Reif, D. M., Martin, M. T., Tan, S. W. et al. (2010). Endocrine profiling and prioritization of environmental chemicals using ToxCast data. Environ Health Perspect 118, 1714-1720. doi:10.1289/ehp.1002180
    • (2010) Environ Health Perspect , vol.118 , pp. 1714-1720
    • Reif, D. M.1    Martin, M. T.2    Tan, S. W.3
  • 265
    • 84873284721 scopus 로고    scopus 로고
    • ToxPi GUI: An interactive visualization tool for transparent integration of data from diverse sources of evidence
    • Reif, D. M., Sypa, M., Lock, E. F. et al. (2013). ToxPi GUI: An interactive visualization tool for transparent integration of data from diverse sources of evidence. Bioinformatics 29, 402-403. doi:10.1093/bioinformatics/bts686
    • (2013) Bioinformatics , vol.29 , pp. 402-403
    • Reif, D. M.1    Sypa, M.2    Lock, E. F.3
  • 266
    • 84934783723 scopus 로고    scopus 로고
    • High-throughput characterization of chemical-associated embryonic behavioral changes predicts teratogenic outcomes
    • Reif, D., Truong, L., Mandrell, D. et al. (2016). High-throughput characterization of chemical-associated embryonic behavioral changes predicts teratogenic outcomes. Arch Toxicol 90, 1459-1470. doi:10.1007/s00204-015-1554-1
    • (2016) Arch Toxicol , vol.90 , pp. 1459-1470
    • Reif, D.1    Truong, L.2    Mandrell, D.3
  • 267
    • 84940606580 scopus 로고    scopus 로고
    • A transcriptome-based classifier to identify developmental toxicants by stem cell testing: Design, validation and optimization for histone deacetylase inhibitors
    • Rempel, E., Hölting, L., Waldmann, T. et al. (2015). A transcriptome-based classifier to identify developmental toxicants by stem cell testing: Design, validation and optimization for histone deacetylase inhibitors. Arch Toxicol 89, 1599-1618. doi:10.1007/s00204-015-1573-y
    • (2015) Arch Toxicol , vol.89 , pp. 1599-1618
    • Rempel, E.1    Hölting, L.2    Waldmann, T.3
  • 269
    • 84975268086 scopus 로고    scopus 로고
    • Large-scale production of mature neurons from human pluripotent stem cells in a three-dimensional suspension culture system
    • Rigamonti, A., Repetti, G., Sun, C. et al. (2016). Large-scale production of mature neurons from human pluripotent stem cells in a three-dimensional suspension culture system. Stem Cell Reports 6, 93-1008. doi:10.1016/j.stemcr.2016.05.010
    • (2016) Stem Cell Reports , vol.6 , pp. 93-1008
    • Rigamonti, A.1    Repetti, G.2    Sun, C.3
  • 270
    • 83755172909 scopus 로고    scopus 로고
    • In vitro assessment of developmental neurotoxicity: Use of microelectrode arrays to measure functional changes in neuronal network ontogeny
    • Robinette, B. L., Harrill, J. A., Mundy, W. R. and Shafer, T. J. (2011). In vitro assessment of developmental neurotoxicity: Use of microelectrode arrays to measure functional changes in neuronal network ontogeny. Front Neuroeng 4, 1. doi:10.3389/ fneng.2011.00001
    • (2011) Front Neuroeng , vol.4 , pp. 1
    • Robinette, B. L.1    Harrill, J. A.2    Mundy, W. R.3    Shafer, T. J.4
  • 271
    • 84938603406 scopus 로고    scopus 로고
    • st century beyond environmental chemicals
    • st century beyond environmental chemicals. ALTEX 32, 171-181. doi:10.14573/altex.1506201
    • (2015) ALTEX , vol.32 , pp. 171-181
    • Rovida, C.1    Asakura, S.2    Daneshian, M.3
  • 272
    • 2342513503 scopus 로고    scopus 로고
    • Glial specification in the vertebrate neural tube
    • Rowitch, D. H. (2004). Glial specification in the vertebrate neural tube. Nat Rev Neurosci 5, 409-419. doi:10.1038/nrn1389
    • (2004) Nat Rev Neurosci , vol.5 , pp. 409-419
    • Rowitch, D. H.1
  • 273
    • 85054058839 scopus 로고    scopus 로고
    • Adverse outcome pathway on chronic binding of antagonist to N-methyl-D-aspartate receptors (NMDARs) during brain development induces impairment of learning and memory abilities
    • OECD Publishing, Paris
    • Sachana, M., Munn, S. and Bal-Price, A. (2016). Adverse outcome pathway on chronic binding of antagonist to N-methyl-D-aspartate receptors (NMDARs) during brain development induces impairment of learning and memory abilities. OECD Series on Adverse Outcome Pathways No. 5, OECD Publishing, Paris. doi:10.1787/5jlsqs5hcrmq-en
    • (2016) OECD Series on Adverse Outcome Pathways No. 5
    • Sachana, M.1    Munn, S.2    Bal-Price, A.3
  • 274
    • 31544478559 scopus 로고    scopus 로고
    • Contralateral hemimicrencephaly and clinical-pathological correlations in children with hemimegalencephaly
    • Salamon, N., Andres, M., Chute, D. J. et al. (2006). Contralateral hemimicrencephaly and clinical-pathological correlations in children with hemimegalencephaly. Brain 129, 352-365. doi:10.1093/brain/awh681
    • (2006) Brain , vol.129 , pp. 352-365
    • Salamon, N.1    Andres, M.2    Chute, D. J.3
  • 275
    • 84894120143 scopus 로고    scopus 로고
    • Lead induces similar gene expression changes in brains of gestationally exposed adult mice and in neurons differentiated from mouse embryonic stem cells
    • Sànchez-Màrtin, F. J., Fan, X., Lindquist, D. M. et al. (2013). Lead induces similar gene expression changes in brains of gestationally exposed adult mice and in neurons differentiated from mouse embryonic stem cells. PLoS One 8, e80558. doi:10.1371/journal.pone.0080558
    • (2013) PLoS One , vol.8 , pp. e80558
    • Sànchez-Màrtin, F. J.1    Fan, X.2    Lindquist, D. M.3
  • 276
    • 84907169600 scopus 로고    scopus 로고
    • Immediate and delayed effects of subchronic Paraquat exposure during an early differentiation stage in 3D-rat brain cell cultures
    • Sandström von Tobel, J., Zoia, D., Althaus, J. et al. (2014). Immediate and delayed effects of subchronic Paraquat exposure during an early differentiation stage in 3D-rat brain cell cultures. Toxicol Lett 230, 188-197. doi:10.1016/j. toxlet.2014.02.001
    • (2014) Toxicol Lett , vol.230 , pp. 188-197
    • Sandström von Tobel, J.1    Zoia, D.2    Althaus, J.3
  • 277
    • 85018252849 scopus 로고    scopus 로고
    • Potential mechanisms of development-dependent adverse effects of the herbicide paraquat in 3D rat brain cell cultures
    • Sandström, J., Broyer, A., Zoia, D. et al. (2017a). Potential mechanisms of development-dependent adverse effects of the herbicide paraquat in 3D rat brain cell cultures. Neurotoxicology 60, 116-124. doi:10.1016/j.neuro.2017.04.010
    • (2017) Neurotoxicology , vol.60 , pp. 116-124
    • Sandström, J.1    Broyer, A.2    Zoia, D.3
  • 278
    • 85006972234 scopus 로고    scopus 로고
    • Development and characterization of a human embryonic stem cell-derived 3D neural tissue model for neurotoxicity testing
    • Sandström, J., Eggermann, E., Charvet, I. et al. (2017b). Development and characterization of a human embryonic stem cell-derived 3D neural tissue model for neurotoxicity testing. Toxicol In Vitro 38, 124-135. doi:10.1016/j.tiv.2016.10.001
    • (2017) Toxicol In Vitro , vol.38 , pp. 124-135
    • Sandström, J.1    Eggermann, E.2    Charvet, I.3
  • 279
    • 84939492154 scopus 로고    scopus 로고
    • Analysis of 17 neurotransmitters, metabolites and precursors in zebrafish through the life cycle using ultrahigh performance liquid chromatography-tandem mass spectrometry
    • Santos-Fandila, A., Vasquez, E., Barranco, A. et al. (2015). Analysis of 17 neurotransmitters, metabolites and precursors in zebrafish through the life cycle using ultrahigh performance liquid chromatography-tandem mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 1001, 191-201. doi:10.1016/j.jchromb.2015.07.040
    • (2015) J Chromatogr B Analyt Technol Biomed Life Sci , vol.1001 , pp. 191-201
    • Santos-Fandila, A.1    Vasquez, E.2    Barranco, A.3
  • 281
    • 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
  • 282
    • 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
  • 283
    • 84874007223 scopus 로고    scopus 로고
    • Neurogenesis in zebrafish – From embryo to adult
    • Schmidt, R., Strähle, U. and Scholpp, S. (2013). Neurogenesis in zebrafish – From embryo to adult. Neural Dev 8, 3. doi:10.1186/1749-8104-8-3
    • (2013) Neural Dev , vol.8 , pp. 3
    • Schmidt, R.1    Strähle, U.2    Scholpp, S.3
  • 284
    • 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
  • 285
    • 81055127451 scopus 로고    scopus 로고
    • Rapid, complete and large-scale generation of post-mitotic neurons from the human LUHMES cell line
    • Scholz, D., Pöltl, 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    Pöltl, D.2    Genewsky, A.3
  • 286
    • 77951673959 scopus 로고    scopus 로고
    • Polybrominated diphenyl ethers induce developmental neurotoxicity in a human in vitro model: Evidence for endocrine disruption
    • Schreiber, T., Gassmann, K., Götz, C. et al. (2010). Polybrominated diphenyl ethers induce developmental neurotoxicity in a human in vitro model: Evidence for endocrine disruption. Environ Health Perspect 118, 572-578. doi:10.1289/ehp. 0901435
    • (2010) Environ Health Perspect , vol.118 , pp. 572-578
    • Schreiber, T.1    Gassmann, K.2    Götz, C.3
  • 288
    • 84930920752 scopus 로고    scopus 로고
    • Evaluation of drug-induced neurotoxicity based on metabolomics, proteomics and electrical activity measurements in complementary CNS in vitro models
    • Schultz, L., Zurich, M. G., Culot, M. et al. (2015). Evaluation of drug-induced neurotoxicity based on metabolomics, proteomics and electrical activity measurements in complementary CNS in vitro models. Toxicol In Vitro 30, 138-165. doi:10.1016/j.tiv.2015.05.016
    • (2015) Toxicol In Vitro , vol.30 , pp. 138-165
    • Schultz, L.1    Zurich, M. G.2    Culot, M.3
  • 289
    • 84943372486 scopus 로고    scopus 로고
    • Human pluripotent stem cell-derived neural constructs for predicting neural toxicity
    • Schwartz, M. P., Hou, Z., Propson, N. E. et al. (2015). Human pluripotent stem cell-derived neural constructs for predicting neural toxicity. Proc Natl Acad Sci U S A 112, 12516-12521. doi:10.1073/pnas.1516645112
    • (2015) Proc Natl Acad Sci U S A , vol.112 , pp. 12516-12521
    • Schwartz, M. P.1    Hou, Z.2    Propson, N. E.3
  • 290
    • 77953022286 scopus 로고    scopus 로고
    • Locomotor activity in zebrafish embryos: A new method to assess developmental neurotoxicity
    • Selderslaghs, I. W., Hooyberghs, J., De Coen, W. and Witters, H. E. (2010). Locomotor activity in zebrafish embryos: A new method to assess developmental neurotoxicity. Neurotoxicol Teratol 32, 460-471. doi:10.1016/j.ntt.2010.03.002
    • (2010) Neurotoxicol Teratol , vol.32 , pp. 460-471
    • Selderslaghs, I. W.1    Hooyberghs, J.2    De Coen, W.3    Witters, H. E.4
  • 291
    • 84875793321 scopus 로고    scopus 로고
    • Assessment of the developmental neurotoxicity of compounds by measuring locomotor activity in zebrafish embryos and larvae
    • Selderslaghs, I. W., Hooyberghs, J., Blust, R. and Witters, H. E. (2013). Assessment of the developmental neurotoxicity of compounds by measuring locomotor activity in zebrafish embryos and larvae. Neurotoxicol Teratol 37, 44-56. doi:10.1016/j.ntt.2013.01.003
    • (2013) Neurotoxicol Teratol , vol.37 , pp. 44-56
    • Selderslaghs, I. W.1    Hooyberghs, J.2    Blust, R.3    Witters, H. E.4
  • 292
    • 84899080543 scopus 로고    scopus 로고
    • Lead exposure disrupts global DNA methylation in human embryonic stem cells and alters their neuronal differentiation
    • Senut, M. C., Sen, A., Cingolani, P. et al. (2014). Lead exposure disrupts global DNA methylation in human embryonic stem cells and alters their neuronal differentiation. Toxicol Sci 139, 142-161. doi:10.1093/toxsci/kfu028
    • (2014) Toxicol Sci , vol.139 , pp. 142-161
    • Senut, M. C.1    Sen, A.2    Cingolani, P.3
  • 293
    • 39549108854 scopus 로고    scopus 로고
    • The circadian system is a target and modulator of prenatal cocaine effects
    • Shang, E. H. and Zhdanova, I. V. (2007). The circadian system is a target and modulator of prenatal cocaine effects. PLoS One 2, e587, doi:10.1371/journal.pone.0000587
    • (2007) PLoS One , vol.2 , pp. e587
    • Shang, E. H.1    Zhdanova, I. V.2
  • 294
    • 80055051560 scopus 로고    scopus 로고
    • Synapse microarray identification of small molecules that enhance synaptogenesis
    • Shi, P., Scott, M. A., Ghosh, B. et al. (2011). Synapse microarray identification of small molecules that enhance synaptogenesis. Nat Commun 2, 510-519. doi:10.1038/ncomms1518
    • (2011) Nat Commun , vol.2 , pp. 510-519
    • Shi, P.1    Scott, M. A.2    Ghosh, B.3
  • 295
    • 84940605691 scopus 로고    scopus 로고
    • Human pluripotent stem cell based developmental toxicity assays for chemical safety screening and systems biology data generation
    • Shinde, V., Klima, S., Sureshkumar, P. S. et al. (2015). Human pluripotent stem cell based developmental toxicity assays for chemical safety screening and systems biology data generation. J Vis Exp 100, e52333. doi:10.3791/52333
    • (2015) J Vis Exp , vol.100 , pp. e52333
    • Shinde, V.1    Klima, S.2    Sureshkumar, P. S.3
  • 296
    • 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
    • Shinde, V., Hoelting, L., Perumal, S. S. et al. (2016). Definition of transcriptome-based indices for quantitative characterization of chemically disturbed stem cell development – Introduction of the STOP-Toxukn and STOP-Toxukk. Arch Toxicol 91, 839-864. doi:10.1007/s00204-016-1741-8
    • (2016) Arch Toxicol , vol.91 , pp. 839-864
    • Shinde, V.1    Hoelting, L.2    Perumal, S. S.3
  • 297
    • 85019231007 scopus 로고    scopus 로고
    • Transformation of astrocytes to a neuroprotective phenotype by microglia via P2Y1 receptor downregulation
    • Shinozaki, Y., Shibata, K., Yoshida, K. et al. (2017). Transformation of astrocytes to a neuroprotective phenotype by microglia via P2Y1 receptor downregulation. Cell Rep 19, 1151-1164. doi:10.1016/j.celrep.2017.04.047
    • (2017) Cell Rep , vol.19 , pp. 1151-1164
    • Shinozaki, Y.1    Shibata, K.2    Yoshida, K.3
  • 298
    • 84996504468 scopus 로고    scopus 로고
    • The cellular and molecular landscapes of the developing human central nervous system
    • Silbereis, J. C., Pochareddy, S., Zhu, Y. et al. (2016). The cellular and molecular landscapes of the developing human central nervous system. Neuron 89, 248-268. doi:10.1016/j. neuron.2015.12.008
    • (2016) Neuron , vol.89 , pp. 248-268
    • Silbereis, J. C.1    Pochareddy, S.2    Zhu, Y.3
  • 299
    • 84987808287 scopus 로고    scopus 로고
    • Functional metabolic interactions of human neuron-astrocyte 3D in vitro networks
    • Simão, D., Terrasso, A. P., Teixeira, A. P. et al. (2016). Functional metabolic interactions of human neuron-astrocyte 3D in vitro networks. Sci Rep 6, 33285. doi:10.1038/srep33285
    • (2016) Sci Rep , vol.6 , pp. 33285
    • Simão, D.1    Terrasso, A. P.2    Teixeira, A. P.3
  • 300
    • 84940795885 scopus 로고    scopus 로고
    • Ketamine-induced toxicity in neurons differentiated from neural stem cells
    • Slikker, W. Jr., Liu, F., Rainosek, S. W. et al. (2015). Ketamine-induced toxicity in neurons differentiated from neural stem cells. Mol Neurobiol 52, 959-969. doi:10.1007/s12035-015-9248-5
    • (2015) Mol Neurobiol , vol.52 , pp. 959-969
    • Slikker, W.1    Liu, F.2    Rainosek, S. W.3
  • 301
    • 84856456273 scopus 로고    scopus 로고
    • Developmental neurotoxicity of organophosphates targets cell cycle and apoptosis, revealed by transcriptional profiles in vivo and in vitro
    • Slotkin, T. A. and Seidler, F. J. (2012). Developmental neurotoxicity of organophosphates targets cell cycle and apoptosis, revealed by transcriptional profiles in vivo and in vitro. Neurotoxicol Teratol 34, 232-241. doi:10.1016/j.ntt.2011.12.001
    • (2012) Neurotoxicol Teratol , vol.34 , pp. 232-241
    • Slotkin, T. A.1    Seidler, F. J.2
  • 302
    • 0036133288 scopus 로고    scopus 로고
    • Unique morphological features of the proliferative zones and postmitotic compartments of the neural epithelium giving rise to striate and extrastriate cortex in the monkey
    • Smart, I. H., Dehay, C., Giroud, P. et al. (2002). Unique morphological features of the proliferative zones and postmitotic compartments of the neural epithelium giving rise to striate and extrastriate cortex in the monkey. Cereb Cortex 12, 37-53. doi:10.1093/cercor/12.1.37
    • (2002) Cereb Cortex , vol.12 , pp. 37-53
    • Smart, I. H.1    Dehay, C.2    Giroud, P.3
  • 304
    • 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
  • 305
    • 33747155008 scopus 로고    scopus 로고
    • Brain-derived neurotrophic factor and control of synaptic consolidation in the adult brain
    • Soulé, J., Messaoudi, E. and Bramham, C. R. (2006). Brain-derived neurotrophic factor and control of synaptic consolidation in the adult brain. Biochem Soc Trans 34, 600-604. doi:10.1042/BST0340600
    • (2006) Biochem Soc Trans , vol.34 , pp. 600-604
    • Soulé, J.1    Messaoudi, E.2    Bramham, C. R.3
  • 306
    • 85007028250 scopus 로고    scopus 로고
    • Zika virus infection induces mitosis abnormalities and apoptotic cell death of human neural progenitor cells
    • Souza, B. S., Sampaio, G. L., Pereira, C. S. et al. (2016). Zika virus infection induces mitosis abnormalities and apoptotic cell death of human neural progenitor cells. Sci Rep 6, 39775. doi:10.1038/srep39775
    • (2016) Sci Rep , vol.6 , pp. 39775
    • Souza, B. S.1    Sampaio, G. L.2    Pereira, C. S.3
  • 307
    • 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
  • 308
    • 85026505222 scopus 로고    scopus 로고
    • Screening the ToxCast phase II libraries for neuroactivity using cortical neurons grown on multi-well microelectrode array (mwMEA) plates
    • Strickland, J. D., Martin, M., Houck, T. et al. (2017). Screening the ToxCast phase II libraries for neuroactivity using cortical neurons grown on multi-well microelectrode array (mwMEA) plates. Arch Toxicol 92, 487-500. doi:10.1007/s00204-017-2035-5
    • (2017) Arch Toxicol , vol.92 , pp. 487-500
    • Strickland, J. D.1    Martin, M.2    Houck, T.3
  • 309
    • 60249094665 scopus 로고    scopus 로고
    • Hazard assessment of methylmercury toxicity to neuronal induction in embryogenesis using human embryonic stem cells
    • Stummann, T. C., Hareng, L. and Bremer, S. (2009). Hazard assessment of methylmercury toxicity to neuronal induction in embryogenesis using human embryonic stem cells. Toxicology 257, 117-126. doi:10.1016/j.tox.2008.12.018
    • (2009) Toxicology , vol.257 , pp. 117-126
    • Stummann, T. C.1    Hareng, L.2    Bremer, S.3
  • 310
    • 0021440739 scopus 로고
    • Blockers of calcium permeability inhibit neurite extension and formation of neuromuscular synapses in cell culture
    • Suarez-Isla, B. A., Pelto, D. J., Thompson, J. M. and Rapoport, S. I. (1984). Blockers of calcium permeability inhibit neurite extension and formation of neuromuscular synapses in cell culture. Brain Res 316, 263-270. doi:10.1016/0165-3806(84)90311-0
    • (1984) Brain Res , vol.316 , pp. 263-270
    • Suarez-Isla, B. A.1    Pelto, D. J.2    Thompson, J. M.3    Rapoport, S. I.4
  • 311
    • 78951487307 scopus 로고    scopus 로고
    • Neural differentiation from human embryonic stem cells as a tool to study early brain development and the neuroteratogenic effects of ethanol
    • Talens-Visconti, R., Sanchez-Vera, I., Kostic, J. et al. (2011). Neural differentiation from human embryonic stem cells as a tool to study early brain development and the neuroteratogenic effects of ethanol. Stem Cells Dev 20, 327-339. doi:10.1089/ scd.2010.0037
    • (2011) Stem Cells Dev , vol.20 , pp. 327-339
    • Talens-Visconti, R.1    Sanchez-Vera, I.2    Kostic, J.3
  • 312
    • 85012245236 scopus 로고    scopus 로고
    • Mechanistic insight into neurotoxicity induced by developmental insults
    • Tamm, C. and Ceccatelli, S. (2017). Mechanistic insight into neurotoxicity induced by developmental insults. Biochem Biophys Res Commun 482, 408-418. doi:10.1016/j. bbrc.2016.10.087
    • (2017) Biochem Biophys Res Commun , vol.482 , pp. 408-418
    • Tamm, C.1    Ceccatelli, S.2
  • 313
    • 84967328422 scopus 로고    scopus 로고
    • Zika virus infects human cortical neural progenitors and attenuates their growth
    • Tang, H., Hammack, C., Ogden, S. C. et al. (2016). Zika virus infects human cortical neural progenitors and attenuates their growth. Cell Stem Cell 18, 587-590. doi:10.1016/j. stem.2016.02.016
    • (2016) Cell Stem Cell , vol.18 , pp. 587-590
    • Tang, H.1    Hammack, C.2    Ogden, S. C.3
  • 314
    • 84949639174 scopus 로고    scopus 로고
    • Sensitivity of neural stem cell survival, differentiation and neurite outgrowth within 3D hydrogels to environmental heavy metals
    • Tasneem, S., Farrell, K., Lee, M. Y. and Kothapalli, C. R. (2016). Sensitivity of neural stem cell survival, differentiation and neurite outgrowth within 3D hydrogels to environmental heavy metals. Toxicol Lett 242, 9-22. doi:10.1016/j. toxlet.2015.11.021
    • (2016) Toxicol Lett , vol.242 , pp. 9-22
    • Tasneem, S.1    Farrell, K.2    Lee, M. Y.3    Kothapalli, C. R.4
  • 315
    • 85037655885 scopus 로고    scopus 로고
    • An adverse outcome pathway for parkinsonian motor deficits associated with mitochondrial complex I inhibition
    • Terron, A., Bal-Price, A., Paini, A. et al. (2018). An adverse outcome pathway for parkinsonian motor deficits associated with mitochondrial complex I inhibition. Arch Toxicol 92, 41-82. doi:10.1007/s00204-017-2133-4
    • (2018) Arch Toxicol , vol.92 , pp. 41-82
    • Terron, A.1    Bal-Price, A.2    Paini, A.3
  • 316
    • 26944472045 scopus 로고    scopus 로고
    • Developmental toxicity of domoic acid in zebrafish (Danio rerio)
    • Tiedeken, J. A., Ramsdell, J. S. and Ramsdell, A. F. (2005). Developmental toxicity of domoic acid in zebrafish (Danio rerio). Neurotoxicol Teratol 27, 711-717. doi:10.1016/j. ntt.2005.06.013
    • (2005) Neurotoxicol Teratol , vol.27 , pp. 711-717
    • Tiedeken, J. A.1    Ramsdell, J. S.2    Ramsdell, A. F.3
  • 317
    • 84155187502 scopus 로고    scopus 로고
    • Non-dioxin-like polychlorinated biphenyls interfere with neuronal differentiation of embryonic neural stem cells
    • Tofighi, R., Wan Ibrahim, W. N., Rebellato, P. et al. (2011). Non-dioxin-like polychlorinated biphenyls interfere with neuronal differentiation of embryonic neural stem cells. Toxicol Sci 124, 192-201. doi:10.1093/toxsci/kfr221
    • (2011) Toxicol Sci , vol.124 , pp. 192-201
    • Tofighi, R.1    Wan Ibrahim, W. N.2    Rebellato, P.3
  • 318
    • 79551580812 scopus 로고    scopus 로고
    • A novel Akt3 mutation associated with enhanced kinase activity and seizure susceptibility in mice
    • Tokuda, S., Mahaffey C. L, Monks, B. et al. (2011). A novel Akt3 mutation associated with enhanced kinase activity and seizure susceptibility in mice. Hum Mol Genet 20, 988-999. doi:10.1093/hmg/ddq544
    • (2011) Hum Mol Genet , vol.20 , pp. 988-999
    • Tokuda, S.1    Mahaffey, C. L2    Monks, B.3
  • 319
    • 84922433050 scopus 로고    scopus 로고
    • Applying adverse outcome pathways (AOPs) to support Integrated approaches to testing and assessment (IATA)
    • Tollefsen, K. E., Scholz, S., Cronin, M. T. et al. (2014). Applying adverse outcome pathways (AOPs) to support Integrated approaches to testing and assessment (IATA). Regul Toxicol Pharmacol 70, 629-640. doi:10.1016/j.yrtph.2014.09.009
    • (2014) Regul Toxicol Pharmacol , vol.70 , pp. 629-640
    • Tollefsen, K. E.1    Scholz, S.2    Cronin, M. T.3
  • 320
    • 84878066293 scopus 로고    scopus 로고
    • MCT8 deficiency: Extrapyramidal symptoms and delayed myelination as prominent features
    • Tonduti, D., Vanderver, A., Berardinelli, A. et al. (2013). MCT8 deficiency: Extrapyramidal symptoms and delayed myelination as prominent features. J Child Neurol 28, 795-800. doi:10.1177/0883073812450944
    • (2013) J Child Neurol , vol.28 , pp. 795-800
    • Tonduti, D.1    Vanderver, A.2    Berardinelli, A.3
  • 321
    • 84937855440 scopus 로고    scopus 로고
    • An adverse outcome pathway framework for neural tube and axial defects mediated by modulation of retinoic acid homeostasis
    • Tonk, E. C., Pennings, J. L. and Piersma, A. H. (2015). An adverse outcome pathway framework for neural tube and axial defects mediated by modulation of retinoic acid homeostasis. Reprod Toxicol 55, 104-113. doi:10.1016/j.reprotox.2014.10.008
    • (2015) Reprod Toxicol , vol.55 , pp. 104-113
    • Tonk, E. C.1    Pennings, J. L.2    Piersma, A. H.3
  • 323
    • 23144449547 scopus 로고    scopus 로고
    • Essential role of protein kinase B gamma (PKB gamma/Akt3) in postnatal brain development but not in glucose homeostasis
    • Tschopp, O., Yang, Z. Z. and Brodbeck, D. (2005). Essential role of protein kinase B gamma (PKB gamma/Akt3) in postnatal brain development but not in glucose homeostasis. Development 132, 2943-2954. doi:10.1242/dev.01864
    • (2005) Development , vol.132 , pp. 2943-2954
    • Tschopp, O.1    Yang, Z. Z.2    Brodbeck, D.3
  • 324
    • 84865552179 scopus 로고    scopus 로고
    • Developmental neurotoxicity guideline study: Issues with methodology, evaluation and regulation
    • Tsuji, R. and Crofton K. M. (2012). Developmental neurotoxicity guideline study: Issues with methodology, evaluation and regulation. Congenit Anom (Kyoto) 52, 122-128. doi:10.1111/ j.1741-4520.2012.00374.x
    • (2012) Congenit Anom (Kyoto) , vol.52 , pp. 122-128
    • Tsuji, R.1    Crofton, K. M.2
  • 325
    • 84979261641 scopus 로고    scopus 로고
    • Is the time right for in vitro neurotoxicity testing using human iPSC-derived neurons?
    • Tukker, A. M., de Groot, M. W., Wijnolts, F. M. et al. (2016). Is the time right for in vitro neurotoxicity testing using human iPSC-derived neurons? ALTEX 33, 261-271. doi:10.14573/ altex.1510091
    • (2016) ALTEX , vol.33 , pp. 261-271
    • Tukker, A. M.1    de Groot, M. W.2    Wijnolts, F. M.3
  • 326
    • 0036742611 scopus 로고    scopus 로고
    • From acquisition to consolidation: On the role of brain-derived neurotrophic factor signaling in hippocampal-dependent learning
    • Tyler, W. J., Alonso, M., Bramham, C. R. and Pozzo-Miller, L. D. (2002). From acquisition to consolidation: On the role of brain-derived neurotrophic factor signaling in hippocampal-dependent learning. Learn Mem 9, 224-237. doi:10.1101/ lm.51202
    • (2002) Learn Mem , vol.9 , pp. 224-237
    • Tyler, W. J.1    Alonso, M.2    Bramham, C. R.3    Pozzo-Miller, L. D.4
  • 327
    • 84923053929 scopus 로고    scopus 로고
    • Assessing skin sensitization hazard in mice and men using non-animal test methods
    • Urbisch, D., Mehling, A., Guth, K. et al. (2015). Assessing skin sensitization hazard in mice and men using non-animal test methods. Regul Toxicol Pharmacol 71, 337-351. doi:10.1016/j. yrtph.2014.12.008
    • (2015) Regul Toxicol Pharmacol , vol.71 , pp. 337-351
    • Urbisch, D.1    Mehling, A.2    Guth, K.3
  • 328
    • 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
  • 329
    • 84962407428 scopus 로고    scopus 로고
    • A multi-laboratory evaluation of microelectrode array-based measurements of neural network activity for acute neurotoxicity testing
    • Vassallo, A., Chiappalone, M., De Camargos, L. R. et al. (2017). A multi-laboratory evaluation of microelectrode array-based measurements of neural network activity for acute neurotoxicity testing. Neurotoxicology 60, 280-292. doi:10.1016/j. neuro.2016.03.019
    • (2017) Neurotoxicology , vol.60 , pp. 280-292
    • Vassallo, A.1    Chiappalone, M.2    De Camargos, L. R.3
  • 330
    • 67349106739 scopus 로고    scopus 로고
    • Neonatal ontogeny and neurotoxic effect of decabrominated diphenyl ether (PBDE 209) on levels of synaptophysin and tau
    • Viberg, H. (2009). Neonatal ontogeny and neurotoxic effect of decabrominated diphenyl ether (PBDE 209) on levels of synaptophysin and tau. Int J Dev Neurosci 27, 423-429. doi:10.1016/j.ijdevneu.2009.05.007
    • (2009) Int J Dev Neurosci , vol.27 , pp. 423-429
    • Viberg, H.1
  • 331
    • 84884179017 scopus 로고    scopus 로고
    • The adverse outcome pathway concept: A pragmatic tool in toxicology
    • Vinken, M. (2013). The adverse outcome pathway concept: A pragmatic tool in toxicology. Toxicology 312, 158-165. doi:10.1016/j.tox.2013.08.011
    • (2013) Toxicology , vol.312 , pp. 158-165
    • Vinken, M.1
  • 332
    • 84867231149 scopus 로고    scopus 로고
    • Neural differentiation of mouse embryonic stem cells as a tool to assess developmental neurotoxicity in vitro
    • Visan, A., Hayess, K., Sittner, D. et al. (2012). Neural differentiation of mouse embryonic stem cells as a tool to assess developmental neurotoxicity in vitro. Neurotoxicology 33, 1135-1146. doi:/10.1016/j.neuro.2012.06.006
    • (2012) Neurotoxicology , vol.33 , pp. 1135-1146
    • Visan, A.1    Hayess, K.2    Sittner, D.3
  • 333
    • 0032810012 scopus 로고    scopus 로고
    • ATP controls neuronal apoptosis triggered by microtubule breakdown or potassium deprivation
    • Volbracht, C., Leist, M. and Nicotera, P. (1999). ATP controls neuronal apoptosis triggered by microtubule breakdown or potassium deprivation. Mol Med 5, 477-489.
    • (1999) Mol Med , vol.5 , pp. 477-489
    • Volbracht, C.1    Leist, M.2    Nicotera, P.3
  • 334
    • 84949225866 scopus 로고    scopus 로고
    • Assessment of learning, memory, and attention in developmental neurotoxicity regulatory studies: Synthesis, commentary, and recommendations
    • Vorhees, C. V. and Makris, S. L. (2015). Assessment of learning, memory, and attention in developmental neurotoxicity regulatory studies: Synthesis, commentary, and recommendations. Neurotoxicol Teratol 52, 109-115. doi:10.1016/j. ntt.2015.10.004
    • (2015) Neurotoxicol Teratol , vol.52 , pp. 109-115
    • Vorhees, C. V.1    Makris, S. L.2
  • 335
    • 33645337367 scopus 로고    scopus 로고
    • An extremely rich repertoire of bursting patterns during the development of cortical cultures
    • Wagenaar, D. A., Pine, J. and Potter, S. M. (2006). An extremely rich repertoire of bursting patterns during the development of cortical cultures. BMC Neurosci 7, 11. doi:10.1186/1471-2202-7-11
    • (2006) BMC Neurosci , vol.7 , pp. 11
    • Wagenaar, D. A.1    Pine, J.2    Potter, S. M.3
  • 336
    • 84896349339 scopus 로고    scopus 로고
    • Design principles of concentration-dependent transcriptome deviations in drug-exposed differentiating stem cells
    • Waldmann, T., Rempel, E., Balmer, N. V. et al. (2014). Design principles of concentration-dependent transcriptome deviations in drug-exposed differentiating stem cells. Chem Res Toxicol 27, 408-420. doi:10.1021/tx400402j
    • (2014) Chem Res Toxicol , vol.27 , pp. 408-420
    • Waldmann, T.1    Rempel, E.2    Balmer, N. V.3
  • 337
    • 85017662944 scopus 로고    scopus 로고
    • Stem cell transcriptome responses and corresponding biomarkers that indicate the transition from adaptive responses to cytotoxicity
    • Waldmann, T., Grinberg, M., König, A. et al. (2017). Stem cell transcriptome responses and corresponding biomarkers that indicate the transition from adaptive responses to cytotoxicity. Chem Res Toxicol 30, 905-922. doi:10.1021/acs.chemrestox. 6b00259
    • (2017) Chem Res Toxicol , vol.30 , pp. 905-922
    • Waldmann, T.1    Grinberg, M.2    König, A.3
  • 338
    • 84930945887 scopus 로고    scopus 로고
    • A multiplexed assay for determination of neurotoxicant effects on spontaneous network activity and viability from microelectrode arrays
    • Wallace, K., Strickland, J. D., Valdivia, P. et al. (2015). A multiplexed assay for determination of neurotoxicant effects on spontaneous network activity and viability from microelectrode arrays. Neurotoxicology 49, 79-85. doi:10.1016/j. neuro.2015.05.007
    • (2015) Neurotoxicology , vol.49 , pp. 79-85
    • Wallace, K.1    Strickland, J. D.2    Valdivia, P.3
  • 339
    • 84923193587 scopus 로고    scopus 로고
    • Wnt signaling pathway participates in valproic acid-induced neuronal differentiation of neural stem cells
    • Wang, L., Liu, Y., Li, S. et al. (2015). Wnt signaling pathway participates in valproic acid-induced neuronal differentiation of neural stem cells. Int J Clin Exp Pathol 8, 578-585.
    • (2015) Int J Clin Exp Pathol , vol.8 , pp. 578-585
    • Wang, L.1    Liu, Y.2    Li, S.3
  • 340
    • 85007227814 scopus 로고    scopus 로고
    • Brain development and Akt signaling: The crossroads of signaling pathway and neurodevelopmental diseases
    • Wang, L., Zhou, K., Fu, Z. et al. (2017). Brain development and Akt signaling: The crossroads of signaling pathway and neurodevelopmental diseases. J Mol Neurosci 61, 379-384. doi:10.1007/s12031-016-0872-y
    • (2017) J Mol Neurosci , vol.61 , pp. 379-384
    • Wang, L.1    Zhou, K.2    Fu, Z.3
  • 341
    • 84860775030 scopus 로고    scopus 로고
    • Extensive transcriptional regulation of chromatin modifiers during human neurodevelopment
    • Weng, M., Zimmer, B., Pöltl, D. et al. (2012). Extensive transcriptional regulation of chromatin modifiers during human neurodevelopment. PLoS One 7, e36708. doi:10.1371/journal. pone.0036708
    • (2012) PLoS One , vol.7 , pp. e36708
    • Weng, M.1    Zimmer, B.2    Pöltl, D.3
  • 342
    • 84904675460 scopus 로고    scopus 로고
    • Lineage-specific regulation of epigenetic modifier genes in human liver and brain
    • Weng, M. K., Nataraj, K., Scholz, D. et al. (2014). Lineage-specific regulation of epigenetic modifier genes in human liver and brain. PLoS One 9, e102035. doi:10.1371/journal. pone.0102035
    • (2014) PLoS One , vol.9 , pp. e102035
    • Weng, M. K.1    Nataraj, K.2    Scholz, D.3
  • 343
    • 84930085761 scopus 로고    scopus 로고
    • Quantitative in vitro-to-in vivo extrapolation in a high-throughput environment
    • Wetmore, B. A. (2015). Quantitative in vitro-to-in vivo extrapolation in a high-throughput environment. Toxicology 332, 94-101. doi:10.1016/j.tox.2014.05.012
    • (2015) Toxicology , vol.332 , pp. 94-101
    • Wetmore, B. A.1
  • 344
    • 84899132272 scopus 로고    scopus 로고
    • Multiparametric high content analysis for assessment of neurotoxicity in differentiated neuronal cell lines and human embryonic stem cell-derived neurons
    • Wilson, M. S., Graham, J. R. and Ball, A. J. (2014). Multiparametric high content analysis for assessment of neurotoxicity in differentiated neuronal cell lines and human embryonic stem cell-derived neurons. Neurotoxicology 42, 33-48. doi:10.1016/j.neuro.2014.03.013
    • (2014) Neurotoxicology , vol.42 , pp. 33-48
    • Wilson, M. S.1    Graham, J. R.2    Ball, A. J.3
  • 345
    • 84876535935 scopus 로고    scopus 로고
    • Modeling transformations of neurodevelopmental sequences across mammalian species
    • Workman, A. D., Charvet, C. J., Clancy, B. et al. (2013). Modeling transformations of neurodevelopmental sequences across mammalian species. J Neurosci 33, 7368-7383. doi:10.1523/ JNEUROSCI.5746-12.2013
    • (2013) J Neurosci , vol.33 , pp. 7368-7383
    • Workman, A. D.1    Charvet, C. J.2    Clancy, B.3
  • 346
    • 84988934809 scopus 로고    scopus 로고
    • Integrated approaches to testing and assessment
    • Worth, A. P. and Patlewicz, G. (2016). Integrated approaches to testing and assessment. Adv Exp Med Biol 856, 317-342. doi:10.1007/978-3-319-33826-2_13
    • (2016) Adv Exp Med Biol , vol.856 , pp. 317-342
    • Worth, A. P.1    Patlewicz, G.2
  • 347
    • 76249090489 scopus 로고    scopus 로고
    • BioGPS: An extensible and customizable portal for querying and organizing gene annotation resources
    • Wu, C., Orozco, C., Boyer, J. et al. (2009). BioGPS: An extensible and customizable portal for querying and organizing gene annotation resources. Genome Biol 10, R130. doi:10.1186/ gb-2009-10-11-r130
    • (2009) Genome Biol , vol.10 , pp. R130
    • Wu, C.1    Orozco, C.2    Boyer, J.3
  • 348
    • 84864521556 scopus 로고    scopus 로고
    • An in vitro method to study the effects of thyroid hormone-disrupting chemicals on neuronal development
    • Xiong, Y., Ibhazehiebo, K., Iwasaki, T. and Koibuchi, N. (2012). An in vitro method to study the effects of thyroid hormone-disrupting chemicals on neuronal development. Neurotoxicology 33, 753-757. doi:10.1016/j.neuro.2012.04.021
    • (2012) Neurotoxicology , vol.33 , pp. 753-757
    • Xiong, Y.1    Ibhazehiebo, K.2    Iwasaki, T.3    Koibuchi, N.4
  • 349
    • 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
  • 350
    • 84996542524 scopus 로고    scopus 로고
    • Unexpected neuroprotective effects of loganin on 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced neurotoxicity and cell death in zebrafish
    • 10.1002/jcb.25749 Yoon, M., Campbell, J. L., Andersen, M. E. and Clewell, H. J. (2012). Quantitative in vitro to in vivo extrapolation of cell-based toxicity assay results. Crit Rev Toxicol 42, 633-652
    • Yao, L., Peng, S. X., Xu, Y. D. et al. (2017). Unexpected neuroprotective effects of loganin on 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced neurotoxicity and cell death in zebrafish. J Cell Biochem 118, 615-628. doi:10.1002/jcb.25749 Yoon, M., Campbell, J. L., Andersen, M. E. and Clewell, H. J. (2012). Quantitative in vitro to in vivo extrapolation of cell-based toxicity assay results. Crit Rev Toxicol 42, 633-652. doi: 10.3109/10408444.2012.692115
    • (2017) J Cell Biochem , vol.118 , pp. 615-628
    • Yao, L.1    Peng, S. X.2    Xu, Y. D.3
  • 351
    • 30344445351 scopus 로고    scopus 로고
    • Refining the effects observed in a developmental neurobehavioral study of ammonium perchlorate administered orally in drinking water to rats. II. Behavioral and neurodevelopment effects
    • York, R. G., Barnett, J., Girard, M. F. et al. (2005). Refining the effects observed in a developmental neurobehavioral study of ammonium perchlorate administered orally in drinking water to rats. II. Behavioral and neurodevelopment effects. Int J Toxicol 24, 451-467. doi:10.1080/10915810500367094
    • (2005) Int J Toxicol , vol.24 , pp. 451-467
    • York, R. G.1    Barnett, J.2    Girard, M. F.3
  • 352
    • 84984825559 scopus 로고    scopus 로고
    • Proliferation, survival and metabolism: The role of PI3K/AKT/mTOR signalling in pluripotency and cell fate determination
    • Yu, J. S. and Cui, W. (2016). Proliferation, survival and metabolism: The role of PI3K/AKT/mTOR signalling in pluripotency and cell fate determination. Development 143, 3050-3060. doi:10.1242/dev.137075
    • (2016) Development , vol.143 , pp. 3050-3060
    • Yu, J. S.1    Cui, W.2
  • 353
    • 84994527381 scopus 로고    scopus 로고
    • Evaluation of the rotenone-induced activation of the Nrf2 pathway in a neuronal model derived from human induced pluripotent stem cells
    • Zagoura, D., Canovas-Jorda, D., Pistollato, F. et al. (2017). Evaluation of the rotenone-induced activation of the Nrf2 pathway in a neuronal model derived from human induced pluripotent stem cells. Neurochem Int 106, 62-73. doi:10.1016/j.neuint.2016.09.004
    • (2017) Neurochem Int , vol.106 , pp. 62-73
    • Zagoura, D.1    Canovas-Jorda, D.2    Pistollato, F.3
  • 354
    • 79955559698 scopus 로고    scopus 로고
    • Latanoprost promotes neurite outgrowth in differentiated RGC-5 cells via the PI3KAkt-mTOR signaling pathway
    • Zheng, J., Feng, X., Hou, L. et al. (2011). Latanoprost promotes neurite outgrowth in differentiated RGC-5 cells via the PI3KAkt-mTOR signaling pathway. Cell Mol Neurobiol 31, 597-604. doi:10.1007/s10571-011-9653-x
    • (2011) Cell Mol Neurobiol , vol.31 , pp. 597-604
    • Zheng, J.1    Feng, X.2    Hou, L.3
  • 355
    • 84942093634 scopus 로고    scopus 로고
    • Inhibitory effect of arsenic trioxide on neuronal migration in vitro and its potential molecular mechanism
    • Zhou, H., Liu, Y., Tan, X. J. et al. (2015). Inhibitory effect of arsenic trioxide on neuronal migration in vitro and its potential molecular mechanism. Environ Toxicol Pharmacol 40, 671-677. doi:10.1016/j.etap.2015.08.026
    • (2015) Environ Toxicol Pharmacol , vol.40 , pp. 671-677
    • Zhou, H.1    Liu, Y.2    Tan, X. J.3
  • 356
    • 84868198811 scopus 로고    scopus 로고
    • PTEN signaling in autism spectrum disorders
    • Zhou, J. and Parada L. F. (2012). PTEN signaling in autism spectrum disorders. Curr Opin Neurobiol 22, 873-879. doi:10.1016/j.conb.2012.05.004
    • (2012) Curr Opin Neurobiol , vol.22 , pp. 873-879
    • Zhou, J.1    Parada, L. F.2
  • 357
    • 85010840300 scopus 로고    scopus 로고
    • Fluoride and arsenic exposure affects spatial memory and activates the ERK/CREB signaling pathway in offspring rats
    • Zhu, Y. P., Xi, S. H., Li, M. Y. et al. (2017). Fluoride and arsenic exposure affects spatial memory and activates the ERK/CREB signaling pathway in offspring rats. Neurotoxicology 59, 56-64. doi:10.1016/j.neuro.2017.01.006
    • (2017) Neurotoxicology , vol.59 , pp. 56-64
    • Zhu, Y. P.1    Xi, S. H.2    Li, M. Y.3
  • 358
    • 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
  • 359
    • 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 12, 357-367. doi:10.1093/toxsci/kfr054
    • (2011) Toxicol Sci , vol.12 , pp. 357-367
    • Zimmer, B.1    Schildknecht, S.2    Kuegler, P. B.3
  • 360
    • 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
  • 361
    • 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
  • 362
    • 84945283142 scopus 로고    scopus 로고
    • Embryological exposure to valproic acid induces social interaction deficits in zebrafish (Danio rerio): A developmental behavior analysis
    • Zimmermann, F. F., Gaspary, K. V., Leite, C. E. et al. (2015). Embryological exposure to valproic acid induces social interaction deficits in zebrafish (Danio rerio): A developmental behavior analysis. Neurotoxicol Teratol 52, 36-41. doi:10.1016/j. ntt.2015.10.002
    • (2015) Neurotoxicol Teratol , vol.52 , pp. 36-41
    • Zimmermann, F. F.1    Gaspary, K. V.2    Leite, C. E.3
  • 363
    • 61449084452 scopus 로고    scopus 로고
    • Acute administration of tributyltin and trimethyltin modulate glutamate and N-methyl-D-aspartate receptor signalling pathway in Sebastiscus marmoratus
    • Zuo, Z., Cai, J., Wang, X. et al. (2009). Acute administration of tributyltin and trimethyltin modulate glutamate and N-methyl-D-aspartate receptor signalling pathway in Sebastiscus marmoratus. Aquat Toxicol 92, 44-49. doi:10.1016/j. aquatox.2009.01.008
    • (2009) Aquat Toxicol , vol.92 , pp. 44-49
    • Zuo, Z.1    Cai, J.2    Wang, X.3
  • 364
    • 0033804109 scopus 로고    scopus 로고
    • Use of aggregating brain cell cultures to study developmental effects of organophosphorus insecticides
    • Zurich, M. G., Honegger, P., Schilter, B. et al. (2000). Use of aggregating brain cell cultures to study developmental effects of organophosphorus insecticides. Neurotoxicology 21, 599-605.
    • (2000) Neurotoxicology , vol.21 , pp. 599-605
    • Zurich, M. G.1    Honegger, P.2    Schilter, B.3
  • 365
    • 0036786148 scopus 로고    scopus 로고
    • Maturation-dependent neurotoxicity of lead aceate in vitro: Implication of glial reactions
    • Zurich, M. G., Eskes, C., Honegger, P. et al. (2002). Maturation-dependent neurotoxicity of lead aceate in vitro: Implication of glial reactions. J Neurosc Res 70, 108-116. doi:10.1002/ jnr.10367
    • (2002) J Neurosc Res , vol.70 , pp. 108-116
    • Zurich, M. G.1    Eskes, C.2    Honegger, P.3
  • 366
    • 84876726626 scopus 로고    scopus 로고
    • Evaluation of aggregating brain cell cultures for the detection of acute organ-specific toxicity
    • Zurich, M. G., Stanzel, S., Kopp-Schneuder, A. et al. (2012). Evaluation of aggregating brain cell cultures for the detection of acute organ-specific toxicity. Toxicol In Vitro 27, 1416-1424. doi:10.1016/j.tiv.2012.06.018
    • (2012) Toxicol In Vitro , vol.27 , pp. 1416-1424
    • Zurich, M. G.1    Stanzel, S.2    Kopp-Schneuder, A.3


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.