Skip to main content Accessibility help
×
Hostname: page-component-77c89778f8-gq7q9 Total loading time: 0 Render date: 2024-07-20T11:14:29.106Z Has data issue: false hasContentIssue false

8 - Predictive models for neurotoxicity assessment

from I - SPECIFIC AREAS OF PREDICTIVE TOXICOLOGY

Published online by Cambridge University Press:  06 December 2010

Jinghai J. Xu
Affiliation:
Merck Research Laboratory, New Jersey
Laszlo Urban
Affiliation:
Novartis Institutes for Biomedical Research, Massachusetts
Get access

Summary

INTRODUCTION

The human nervous system is one of the most complex organ systems in terms of both structure and function. It contains billions of neurons, each forming thousands of synapses leading to a very large number of connections. It also contains perhaps ten times more glial cells (astrocytes, oligodendrocytes, microglia) than neurons, which play important roles in the overall development and functioning of the nervous system. Anatomically, the nervous system is composed of a central (CNS) and a peripheral (PNS) component, whose basic functions are to detect and relay sensory information inside and outside the body, to direct motor functions, and to integrate thought processes, learning, and memory. Such functions and their complexity, together with some intrinsic characteristics (e.g., mature neurons do not divide, they are highly dependent upon oxygen and glucose) make the nervous system particularly vulnerable to toxic insults.

Neurotoxicity can be defined as any adverse effect on the chemistry, structure, or function of the nervous system, during development or at maturity, induced by chemical or physical influences. A first issue is what constitutes an adverse effect. A proposed definition of an adverse effect is “any treatment related change which interferes with normal function and compromises adaptation to the environment.” Thus, most morphological changes such as neuronopathy (a loss of neurons), axonopathy (a degeneration of the neuronal axon), or myelinopathy (a loss of the glial cells surrounding the axon), or other gliopathies, would be considered adverse, even if structural and/or functional changes were mild or transitory.

Type
Chapter
Information
Publisher: Cambridge University Press
Print publication year: 2010

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Barres, BA. The mystery and magic of glia: A perspective on their roles in health and disease. Neuron. 2008;60:430–440.CrossRefGoogle ScholarPubMed
Costa, LG. Neurotoxicity testing: A discussion of in vitro alternatives. Environ Health Perspect. 1998;106(Suppl. 2) 505–510.CrossRefGoogle ScholarPubMed
,ECETOC. Evaluation of the Neurotoxic Potential of Chemicals. Brussels: European Center for Ecotoxicology and Toxicology of Chemicals; 1992.Google Scholar
Grandjean, P, Landrigan, PJ. Developmental neurotoxicity of industrial chemicals. Lancet. 2006; 368: 2167–2178.CrossRefGoogle ScholarPubMed
Spencer, PS, Schaumburg, HH, Ludolph, AC, eds. Experimental and Clinical Neurotoxicology. Oxford: Oxford University Press: 2000:1310.Google Scholar
Jensen, KF, Catalano, SM. Brain morphogenesis and developmental neurotoxicology. In: Slikker, W, Chang, LW, eds., Handbook of Developmental Neurotoxicology. San Diego: Academic Press; 1998:3–41.Google Scholar
Costa, LG, Giordano, G. Developmental neurotoxicity of polybrominated diphenyl ether (PBDE) flame retardants. Neurotoxicology. 2007;28:1047–1067.CrossRefGoogle ScholarPubMed
Crofton, KM. Thyroid disrupting chemicals: mechanisms and mixtures. Int. J. Androl. 2008;31:209–223.CrossRefGoogle ScholarPubMed
,USEPA (U.S. Environmental Protection Agency). Health Effects Test Guidelines. OPPTS 870.6200. Neurotoxicity screening battery. Washington, DC: USEPA; 1998.
,OECD (Organization for Economic Co-operation and Development). Test Guideline 424. OECD Guideline for Testing of Chemicals. Neurotoxicity study in rodents. Paris: OECD; 1997.
Makris, SL, Raffaele, K, Allen, S, et al. A retrospective performance assessment of the developmental neurotoxicity study in support of OECD test guideline 426. Environ. Health Perspect. 2009;117:17–25.CrossRefGoogle ScholarPubMed
,USEPA (U.S. Environmental Protection Agency). Health Effects Test Guidelines. OPPTS 870.6300. Developmental neurotoxicity study. Washington, DC: USEPA; 1998.
,OECD (Organization for Economic Co-operation and Development). Test Guideline 426. OECD Guideline for Testing of Chemicals. Developmental neurotoxicity study. Paris: OECD; 2007.
Claudio, L, Kwa, WC, Russell, AL, et al. Testing methods for developmental neurotoxicity of environmental chemicals. Toxicol Appl Pharmacol. 2000;164:1–14.CrossRefGoogle ScholarPubMed
Cory-Slechta, DA, Crofton, KM, Foran, JA, et al. Methods to identify and characterize developmental neurotoxicity for human health risk assessment. I: Behavioral effects. Environ Health Perspect. 109(Suppl. 1):79–91.CrossRef
Harry, GJ, Billingsley, M, Bruinink, A, et al. In vitro techniques for the assessment of neurotoxicity. Environ Health Perspect. 1998;106(Suppl. 1):131–158.CrossRefGoogle ScholarPubMed
Sunol, C, Babot, Z, Fonfria, E, et al. Studies with neuronal cells: From basic studies of mechanisms of neurotoxicity to the prediction of chemical toxicity. Toxicol In Vitro. 2008;22:1350–1355.CrossRefGoogle ScholarPubMed
Gartlon, J, Kinsner, A, Bal-Price, A, et al. Evaluation of a proposed in vitro test strategy using neuronal and non-neuronal cell systems for detecting neurotoxicity. Toxicol In Vitro. 2006;20:1569–1581.CrossRefGoogle ScholarPubMed
Bal-Price, AK, Hogberg, HT, Buzanska, L, et al. Relevance of in vitro neurotoxicity testing for regulatory requirements: Challenges to be considered. Neurotoxicol. Teratol. 2010;32:36–41.CrossRefGoogle ScholarPubMed
Silva, RFM, Falcao, AS, Fernandes, A, et al. Dissociated primary nerve cell cultures as models for assessment of neurotoxicity. Toxicol Lett. 2006;163:1–9.CrossRefGoogle ScholarPubMed
Coecke, S, Eskes, C, Gartlon, J, et al. The value of alternative testing for neurotoxicity in the context of regulatory needs. Environ Toxicol Pharmacol. 2006;21:153–167.CrossRefGoogle ScholarPubMed
Honegger, P, Monnet-Tschudi, F. Aggregating neural cell cultures. In: (Fedoroff, S, Richardson, A, eds. Protocols for Neural Cell Cultures. Ottawa: Humana Press: 2001:199–218.Google Scholar
McLean, WG, Holme, AD, Janneh, O, et al. The effect of benomyl on neurite outgrowth in mouse NB2A and human SH-SY5Y neuroblastoma cells in vitro. Neurotoxicology. 1998;19:629–632.Google ScholarPubMed
Costa, LG, Fattori, V, Giordano, G, et al. An in vitro approach to assess the toxicity of certain food contaminants: Methylmercury and polychlorinated biphenyls. Toxicology. 2007;237:65–76.CrossRefGoogle Scholar
Dey, S, Mactutus, CF, Booze, RM, et al. Specificity of prenatal cocaine on inhibition of locus coeruleus neurite outgrowth. Neuroscience. 2006;139:899–907.CrossRefGoogle ScholarPubMed
Radio, NM, Mundy, WR. Developmental neurotoxicity testing in vitro: Models for assessing chemical effects on neurite outgrowth. Neurotoxicology. 2008;29:361–376.CrossRefGoogle ScholarPubMed
Tamm, C, Duckworth, J, Hemanson, O, et al. High susceptibility of neural stem cells to methylmercury toxic effects on cell survival and neuronal differentiation. J Neurochem. 2006; 7:69–78.CrossRefGoogle Scholar
Giordano, G, Kavanagh, TJ, Costa, LG. Neurotoxicity of a polybrominated diphenyl ether mixture (DE-71) in mouse neurons and astrocytes is modulated by intracellular glutathione levels. Toxicol Appl Pharmacol. 2008;232:161–168.CrossRefGoogle ScholarPubMed
Giordano, G, White, CC, Mohar, I, et al. Glutathione levels modulate domoic acid- induced apoptosis in mouse cerebellar granule cells. Toxicol. Sci. 2007;100: 433–444.CrossRefGoogle ScholarPubMed
Guizzetti, M, Thompson, BD, Kim, Y, et al. Role of phospholipase D signaling in ethanol induced inhibition of carbachol-stimulated DNA synthesis of 1321N1 astrocytoma cells. J Neurochem. 2004;90:646–653.CrossRefGoogle ScholarPubMed
Kodavanti, PR, Ward, TR. Differential effects of commercial polybrominated diphenyl ether and polychlorinated biphenyl mixtures on intracellular signaling in rat brain in vitro. Toxicol Sci. 2005;85:952–962.CrossRefGoogle ScholarPubMed
Zurich, MG, Honegger, P, Schilter, B, et al. Involvement of glial cells in the neurotoxicity of parathion and chlorpyrifos. Toxico. Sci. 2004;201:97–104.Google ScholarPubMed
Giordano, G, Kavanagh, TJ, Costa, LG. Mouse cerebellar astrocytes protect cerebellar granule neurons against toxicity of the polybrominated diphenyl ether (PBDE) mixture DE-71. Neurotoxicology. 2009;30:326–329.CrossRefGoogle ScholarPubMed
Janigro, D, Costa, LG. Effects of trimethyltin on granule cells excitability in the in vitro rat dentate gyrus. Neurotoxicol Teratol. 1987;9:33–38.CrossRefGoogle ScholarPubMed
Goldoni, M, Vettori, MV, Alinovi, R, et al. Models of neurotoxicity: Extrapolation of threshold doses in vitro. Risk Anal. 2003;23:505–514.CrossRefGoogle Scholar
Lotti, M, Moretto, A. Organophosphate-induced delayed polyneuropathy. Toxicol Rev. 2005;24:37–49.CrossRefGoogle ScholarPubMed
Ehrich, M, Correll, L, Veronesi, B. Acetylcholinesterase and neuropathy target esterase inhibitions in neuroblastoma cells to distinguish organophosphorus compounds causing acute and delayed neurotoxicity. Fund Appl Toxicol. 1997;38:55–63.CrossRefGoogle ScholarPubMed
Breier, JM, Radio, NM, Mundy, WR, et al. Development of a high-throughput screening assay for chemical effects on proliferation and viability of immortalized human neural progenitor cells. Toxicol Sci. 2008;105:119–133.CrossRefGoogle ScholarPubMed
Radio, NM, Breier, JM, Shafer, TJ, et al. Assessment of chemical effects on neurite outgrowth in PC12 cells using high content screening. Toxicol Sci. 2008;105:106–118.CrossRefGoogle ScholarPubMed
Vliet, E, Morath, S, Eskes, C, et al. A novel metabolomics approach for neurotoxicity testing, proof of principle for methylmercury chloride and caffeine. Neurotoxicology. 2008;29:1–12.CrossRefGoogle Scholar
Vliet, E, Stoppini, L, Balestrino, M, et al. Electrophysiological recording of re-aggregating brain cell cultures on multi-electrode arrays to detect acute neurotoxic effects. Neurotoxicology. 2007;28:1136–1146.CrossRefGoogle ScholarPubMed
Peterson, RT, Nass, R, Boyd, WA, et al. Use of non-mammalian alternative models for neurotoxicological study. Neurotoxicology. 2008;29:546–555.CrossRefGoogle ScholarPubMed
Buznikov, GA, Nikitina, , Bezuglov, VV, et al. An invertebrate model of the developmental neurotoxicity of insecticides: Effects of chlorpyrifos and dieldrin in sea urchin embryos and larvae. Environ Health Perspect. 2001;109:651–661.CrossRefGoogle ScholarPubMed
Falugi, C, Lammerding-Koppel, M, Aluigi, MG. Sea urchin development: An alternative model for mechanistic understanding of neurodevelopment and neurotoxicity. Birth Defects Res (Pt C). 2008;84:188–2003.CrossRefGoogle ScholarPubMed
Zon, LJ, Peterson, RT. In vivo drug discovery in the zebrafish. Nature Rev Drug Discov. 2005;4:35–44.CrossRefGoogle ScholarPubMed
Blader, P, Strahle, U. Zebrafish developmental genetics and central nervous system development. Hum Mol Genet. 2000;9:945–951.CrossRefGoogle ScholarPubMed
Jeong, JY, Kwon, HB, Ahn, JC, et al. Functional and developmental analysis of the blood–brain barrier in zebrafish. Brain Res Bull. 2008;75:619–628.CrossRefGoogle ScholarPubMed
Ton, C, Lin, Y, Willett, C. Zebrafish as a model for developmental neurotoxicity testing. Birth Defects Res (Pt. A.) 2006;76:553–567.CrossRefGoogle ScholarPubMed
Parng, C, Roy, NM, Ton, C, et al. Neurotoxicity assessment using zebrafish. J. Pharmacol Toxicol Meth. 2007;55:103–112.CrossRefGoogle ScholarPubMed
McKinley, ET, Baranowski, TC, Blavo, , et al. Neuroprotection of MPTP-induced toxicity in zebrafish dopaminergic neurons. Brain Res Mol. Brain Res. 2005;141:128–137.CrossRefGoogle ScholarPubMed
Kaletta, T, Hengartner, MO. Finding function in novel targets: C. elegans as a model organism. Nat Rev Drug Discovery. 2006;5:387–398.CrossRefGoogle ScholarPubMed
Helmke, KJ, Avila, DS, Aschner, M. Utility of Caenorhabditis elegans in high throughput neurotoxicological research. Neurotoxicol. Teratol. 2010;32:62–67.CrossRefGoogle Scholar
White, JG, Southgate, J, Thomson, JN, et al. The structure of the nervous system of the nematode Caenorhabditis elegans. Philos Trans R Soc Lond, B Bio. Sci. 1986; 314:1–340.CrossRefGoogle ScholarPubMed
Leung, MCK, Williams, PL, Benedetto, A, et al. Caenorhabditis elegans: An emerging model in biomedical and environmental toxicology. Toxicol Sci. 2008;106:5–28.CrossRefGoogle ScholarPubMed
Hengartner, MO, Horvitz, HR. Programmed cell death in Caenorhabditis elegans. Curr Op Genet Dev. 1994;4:581–586.CrossRefGoogle ScholarPubMed
Boyd, WA, Smith, MV, Kissling, G, et al. Medium- and high-throughput screening of neurotoxicants using C. elegans. Neurotoxicol Teratol. 2010;32:68–73.CrossRefGoogle ScholarPubMed
Grosset, KA, Grosset, DG. Prescribed drugs and neurological complications. J Neurol Neurosurg Psychi. 2004;75:2–8.CrossRefGoogle ScholarPubMed
Windebank, AJ, Grisold, W. Chemotherapy-induced neuropathy. J Periph Nervous Sys. 2008;13:27–46.CrossRefGoogle ScholarPubMed
Ferrari, A. Headache: One of the most common and troublesome adverse reactions to drugs. Curr Drug Saf. 2006;1:43–58.CrossRefGoogle ScholarPubMed
Piotrowski, PL, Sumter, BG, Malling, HV, et al. A toxicity evaluation and predictive system based on neural networks and wavelets. J Chem Inf Model. 2007;47:676–685.CrossRefGoogle ScholarPubMed
Lin, JH. CSF as a surrogate for assessing CNS exposure: An industrial perspective. Curr Drug Metab. 2008;9: 6–59.CrossRefGoogle ScholarPubMed
Abbott, NJ, Dolman, , Patabendige, AK. Assays to predict drug permeation across the blood–brain barrier, and distribution to brain. Curr Drug Metab. 2008;9:901–910.CrossRefGoogle Scholar
Vastag, M, Keseru, GM. Current in vitro and in silico models of blood–brain barrier penetration: A practical view. Curr Opin Drug Discov Devel. 2009;12:115–124.Google ScholarPubMed
Hallier-Vanuxeem, D, Prieto, P, Culot, M, et al. New strategy for alerting central nervous system toxicity: integration of blood–brain barrier toxicity and permeability in neurotoxicity assessment. Toxicol In Vitro. 2009;23:447–453.CrossRefGoogle ScholarPubMed
Xu, JJ, Henstock, PV, Dunn, MC, et al. Cellular imaging predictions of clinical drug-induced liver injury. Toxicol Sci. 2008;105:97–105.CrossRefGoogle ScholarPubMed

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×