Skip to main content

Neurodegenerations Induced by Organophosphorous Compounds

  • Chapter
Neurodegenerative Diseases

Part of the book series: Advances in Experimental Medicine and Biology ((AEMB,volume 724))

Abstract

Organophosphorous compounds (OPs) are widely used in agriculture, industry and the home. Though best known for their acute effects when used as pesticides, which target acetylcholinesterase (AChE) activity in neuromuscular junctions and the central nervous system, not all OPs are potent inhibitors of this enzyme. The widespread use of OPs has heightened concern regarding their toxicity in man, with numerous reports linking OPs to various forms of delayed neuropathy encompassing a range of neurodegenerative, psychological and neurobehavioral effects. There is mounting evidence to suggest that sub-acute levels of OPs have the ability to interact directly with a range of target proteins in addition to AChE (i.e., noncholinergic targets), causing major disruption of membrane and protein turnover, protein phosphorylation, mitochondrial dysfunction, oxidative stress and cytoskeletal re-organisation, although the mechanisms involved are not fully understood. However, major advances have been made in the study of one OP binding protein neuropathy target esterase (NTE) in terms of its true physiological role. Additionally, there is increasing evidence for the ability of OPs to cause disruption in a number of metabolic and cell signalling pathways that affect neuronal cell proliferation, differentiation and survival and to interact direct with non-esterase proteins such as tubulin. The aim of this chapter is to review our current understanding of delayed neurotoxicity, to discuss how these molecular events may relate to each other and to suggest possible future directions in mechanistic studies of OP toxicity.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 129.00
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
Hardcover Book
USD 169.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Preview

Unable to display preview. Download preview PDF.

Unable to display preview. Download preview PDF.

References

  1. Chambers JE. Organophosphate compounds an overview. In: Chambers JE, Levi P, eds. Organophosphates: Chemistry, fate and effects. San Diego: Academic Press, 1992:3–17.

    Google Scholar 

  2. Krieger RI, ed. Handbook of pesticide toxicology: Principles, 2nd edition. San Diego: Academic Press, 2001.

    Google Scholar 

  3. Gupta RC, ed. Toxicology of organophosphate and carbamate compounds. San Diego: Elsevier, 2006.

    Google Scholar 

  4. Aldridge WN. Tricresyl phosphates and cholinesterase. Biochem J 1954; 56:185–189.

    PubMed  CAS  Google Scholar 

  5. Olsen KR, ed. Poisoning and drug overdose. Stamford, Connecticut: Appleton and Lange, 1998.

    Google Scholar 

  6. Olsen KR, ed. Poisoning and drug overdose. Stamford Connecticut; Appleton and Lange, 1998.

    Google Scholar 

  7. Committee on Toxicology (COT) of Chemicals in food, consumer products and he environment organophosphates. Department of Health, 2007.

    Google Scholar 

  8. Environmental Protection Agency. Annual Report, 2000. Office of Pesticide Programs. http://www.epa.gov/ oppfead1/annual/2000/2000annual.pdf

    Google Scholar 

  9. Environmental Protection Agency. Reregistration Eligibility Decision for Diazinon, 2006. http://www.sps. gov/pesticides/reregistration/REDS/diazinon_red.pdf

    Google Scholar 

  10. European Commission (EC) decision of 6 June 2007 concerning the non inclusion of diazinon in Annex I to the Council Directive 91/414/EEC and the withdrawal of authorisations for plant protection agents containing that substance. Official Journal of the European Union 2007; 140:9–10.

    Google Scholar 

  11. Australian Pesticides and Veterinary Medicines Authority (APVMA). APVMA suspends the use of diazinon for sheep dipping and jetting. Media Release, 2007.

    Google Scholar 

  12. Department of Primary Industries (DPI) Sheep body lice: Control and eradication. Agriculture Notes of the DPI State of Victoria, 2007; AG1110.

    Google Scholar 

  13. AbouDonia MB, Lapadula DM. Mechanisms of organophosphorusester induced delayed neurotoxicity: Type I and II. Ann Rev Pharmacol Toxicol 1990; 30:405–440.

    Article  CAS  Google Scholar 

  14. Lotti M. The pathogenesis of organophosphate polyneuropathy. Crit Rev Toxicol 1992; 21:465–487.

    Article  CAS  Google Scholar 

  15. AbouDonia MB. Organophosphorus ester induced chronic neurotoxicity. Arch Env Health 2003; 58:484–497.

    Article  CAS  Google Scholar 

  16. Bishop EL, Stewart HC. Incidence of partial paralysis. Am J Public Health 1930; 20:1307–1312.

    Article  CAS  Google Scholar 

  17. Zeligs MA. Upper motor neuron sequelae in “Jake” paralysis: a clinical follow up study. J Nerv Ment Dis 1938; 87:464–470.

    Article  Google Scholar 

  18. Richardson RJ, Moore TB, Kayyali US et al. Chlorpyrifos: Assessment of potential or delayed neurotoxicity by repeated dosing in adult hens with monitoring of brain acetylcholinesterase, brain and lymphocyte neurotoxic esterase and plasma butyrylcholinesterase activities. Toxicol Sci 1993; 21:89–96.

    Article  CAS  Google Scholar 

  19. Chow E, Seiber JN, Wilson BW. Isophenos and an in vitro assay for delayed neuropathic potential. Toxicol Appl Pharmacol 1986; 83:178–183.

    Article  CAS  Google Scholar 

  20. de Blaquiere GE, Waters L, Blain PG et al. Electrophysiological and biochemical effects of single and multiple doses of the organophosphate diazinon in the mouse. Toxicol Appl Pharmacol 2000; 166:81–91.

    Article  PubMed  Google Scholar 

  21. Johnson MK. The primary biochemical lesion leading to the delayed neurotoxic effects of some organophosphorus esters. J Neurochem 1974; 23:785–789.

    Article  PubMed  CAS  Google Scholar 

  22. Johnson MK. The target for initiation of delayed neurotoxicity by organophosphorus esters: biochemical studies and toxicological applications. In: Hodgson E, Bend JR, Philpot RM, Eds. Reviews in Biochemical Toxicology. New York: Elsevier, 1982; 4:141–212.

    Google Scholar 

  23. Johnson MK. Organophosphates and delayed neuropathy-is NTE alive and well? Toxicol Appl Pharmacol 1990; 102:385–389.

    Article  PubMed  CAS  Google Scholar 

  24. Committee on Air Quality in Passenger Cabins of Commercial Aircraft, Board on Environmental Studies and Toxicology, National Research Council, National Academy of Sciences. The Airliner Cabin Environment and Health of Passengers and Crew. Washington DC; National Academic Press, 2002.

    Google Scholar 

  25. Winder C, ed. Contaminated Air Protection: Proceedings of the Air Safety and Cabin Air Quality International Aero Industry Conference. British Airline Pilots Association (BALPA) and the University of New South Wales, Australia, 2005.

    Google Scholar 

  26. Winder C. Air monitoring studies for air cabin contamination. Curr Top Toxicol 2006; 3:33–48.

    Google Scholar 

  27. Civil Aviation Authority. Cabin air quality, CAA paper, 2004:04.

    Google Scholar 

  28. Harris MO, McLure P, Chessin RL et al. Toxicological profile for hydraulic fluids. ATDSR, USA; 1997.

    Google Scholar 

  29. Hanhela PJ, Kibby J, DeNola G et al. Organophosphate and amine contamination of cockpit air in the Hawk, F-111 and Hercules C-130 aircraft. DSTO Defence Science and Technology Organisation, Victoria, Australia; 2005.

    Google Scholar 

  30. Nutley BP, Crocker J. Biological monitoring of workers occupationally exposed to organophosphorous pesticides. Pesticide Science 1993; 39:315–322.

    Article  Google Scholar 

  31. Pilkington A, Buchanan D, Jamal GA et al. An epidemiological study of the relations between exposure to organophosphate pesticides and indices of chronic peripheral neuropathy and neuropsychological abnormalities in sheep farmers and dippers. Occup Environ Med 2001; 58:702–710.

    Article  PubMed  CAS  Google Scholar 

  32. Costa LG. Current issues in organophosphate toxicology. Clin Chim Acta 2006; 366:1–13.

    Article  PubMed  CAS  Google Scholar 

  33. Karalliedde L, Baker D, Marrs TC. Organophosphate-induced intermediate syndrome: aetiology and relationships with myopathy. Toxicol Rev 2006; 25:1–14.

    Article  PubMed  CAS  Google Scholar 

  34. Slotkin TA, Ryde TI, Levin ED et al. Developmental neurotoxicity of low dose diazinon exposure of neonatal rats: Effects on serotonin systems in adolescence and adulthood. Brain Res Bull 2008; 75:640–647.

    Article  PubMed  CAS  Google Scholar 

  35. Slotkin T, Seidler FJ. Developmental neurotoxicants target differentiation into the serotonin phenotype: Chlorpyrifos, diazinon, dieldrin and divalent nickel. Toxicol Appl Pharmacol 2008; 233:211–219.

    Article  PubMed  CAS  Google Scholar 

  36. Thiermann H, Szinicz L, Eyer F et al. Modern strategies in therapy of organophosphate poisoning. Toxicol Lett 1999; 107:233–239.

    Article  PubMed  CAS  Google Scholar 

  37. Eddleston M, Buckley NA, Eyer P et al. Management of acute organophosphorus pesticide poisoning. The Lancet 2008; 371:597–607.

    Article  CAS  Google Scholar 

  38. Akhmedova SN, Yakimovsky AK, Schwartz EI. Paraoxonase 1 Met-Leu polymorphism is associated with Parkinson’s disease. J Neurol Sci 2001; 184:179–182.

    Article  PubMed  CAS  Google Scholar 

  39. Lotti M, Moretto R, Zoppellari R et al. Inhibition of neuropathy target esterase predicts the development of OPIDN. Arch Toxicol 1986; 59:176–179.

    Article  PubMed  CAS  Google Scholar 

  40. Bissbort SH, Vermaak WJH, Elias J et al. Novel test and its automation for the determination of erythrocyte acetylcholinesterase and its application to organophosphate poisoning. Clin Chim Acta 2001; 303:139–145.

    Article  PubMed  CAS  Google Scholar 

  41. Jokanovic M, Kosanovic M, Brkic D et al. Organophosphate induced delayed polyneuropathy in man: an overview. Clin Neurol Neurosurg 2010; in press.doi:10.1016/j.clineuro.2010.08.015

    Google Scholar 

  42. Lotti M, Johnson MK. Neurotoxicity of organophosphorus pesticides: predictions can be based on in vitro studies with hen and human enzymes. Arch Toxicol 1978; 41:215–221.

    Article  PubMed  CAS  Google Scholar 

  43. El Sebae AH, Soliman SA, AboElamayem M et al. Neurotoxicity of organophosphorus insecticides leptophos and EPN. J Env Sci Health 1977; B12:269–288.

    Article  Google Scholar 

  44. Abou-Donia MB, Graham DG. Delayed neurotoxicity of sub chronic oral administration of leptophos to hens: recovery during four months after exposure. J Toxicol Env Health 1979; 5:1133–1147.

    Article  CAS  Google Scholar 

  45. Rosenstock L, Keifer M, Daniell WE et al. Chronic central nervous system effects of acute organophosphate pesticide intoxication. The Lancet 1991; 338:223–227.

    Article  CAS  Google Scholar 

  46. Mackenzie Ross SJ, Brewin CR, Curran HV et al. Neuropsychological and psychiatric functioning in sheep farmers exposed to low levels of organophosphate pesticides. Neuro Toxicol Teratol 2010; 32:452–459.

    Article  CAS  Google Scholar 

  47. Jett DA, Navoa RV, Beckles RA et al. Cognitive function and cholinergic neurochemistry in weanling rats exposed to chlorpyrifos. Toxicol Appl Pharmacol 2001; 174:89–98.

    Article  PubMed  CAS  Google Scholar 

  48. Abdel Rasoul GM, Abou Salem ME, Mechael AA et al. Effects of occupational pesticide exposure on children applying pesticides. Neuro Toxicology 2008; 29:833–888.

    CAS  Google Scholar 

  49. Casida JE, Quistad GB. Organophosphate toxicology: Safety aspects of nonacetylcholinesterase secondary targets. Toxicology 2004; 17:983–998.

    CAS  Google Scholar 

  50. Glynn P. Neuropathy target esterase. Biochem J 1999; 344:625–631.

    Article  PubMed  CAS  Google Scholar 

  51. Glynn P. Neural development and neuropathy target esterase: two faces of neuropathy target esterase. Prog Neurobiol 2000; 61:61–74.

    Article  PubMed  CAS  Google Scholar 

  52. Kropp TJ, Richardson RJ. Relative inhibitory potencies of chlorpyrifosoxon, chlorpyrifos methyl oxon and mipafox for acetylcholinesterase versus neuropathy target esterase. J Toxicol Environ Health A 2003; 278:8820–8825.

    Google Scholar 

  53. Glynn P. Molecular cloning of neuropathy target esterase. Chem Biol Interact 1999; 119–120:513–517.

    Article  PubMed  Google Scholar 

  54. Forshaw PJ, Atkins J, Ray DE et al. The catalytic domain of human neuropathy target esterase mediates an organophosphate sensitive ion conductance across liposome membranes. J Neurochem 2001:400–406.

    Google Scholar 

  55. Atkins J, Luthjens LH, Hom ML et al. Monomers of the catalytic domain of human neuropathy target esterase are active in the presence of phospholipid. Biochem J 2002; 361:119–123.

    Article  PubMed  CAS  Google Scholar 

  56. Li Y, Dinsdale D, Glynn P. Protein domains, catalytic activity and subcellular distribution of neuropathy target esterase in mammalian cells. J Biol Chem 2003; 278:8820–8825.

    Article  PubMed  CAS  Google Scholar 

  57. Glynn P. Neuropathy target esterase and phospholipiddeacylation. Biochim Biophys Acta 2005; 1736:87–93.

    PubMed  CAS  Google Scholar 

  58. Kretzschmar D, Hasan G, Sharma S et al. The Swiss cheese mutant causes glial hyper wrapping and brain degeneration in Drosophila. J Neurosci 1997; 17:7425–7432.

    PubMed  CAS  Google Scholar 

  59. Akassoglou K, Malester B, Xu J et al. Brain-specific degeneration of neuropathy target esterase/swiss cheese results in neurodegeneration. Proc Natl Acad Sci USA 2004; 101:5075–5780.

    Article  PubMed  CAS  Google Scholar 

  60. Mühlig-Versen M, Bettencourt de Cruz A, Tschäpe JA et al. Loss of swiss cheese/neuropathy target esterase activity causes disruption of phosphatidylcholine homeostasis and neuronal and glial cell death in Drosophila. J Neurosci 2005; 25:2865–2873.

    Article  PubMed  Google Scholar 

  61. Lush MJ, Li Y, Read DJ et al. Neuropathy target esterase and a homologous Drosophila neurodegeneration mutant protein contain a domain conserved from bacteria to man. Biochem J 1998; 332:1–4.

    PubMed  CAS  Google Scholar 

  62. Rainier S, Bui M, Mark E et al. Neuropathy target esterase gene mutations cause motor neuron disease. Am J Human Genet 2008; 82:780–785.

    Article  CAS  Google Scholar 

  63. Hein ND, Stuckley JA, Rainier SR et al. Constructs of human neuropathy target esterase catalytic domain containing mutations related to motor neuron disease have altered enzymatic properties. Toxicol Lett 2010; 196:67–73.

    Article  PubMed  CAS  Google Scholar 

  64. Read DJ, Li Y, Chao MV et al. Neuropathy target esterase is required for adult vertebrate axon maintenance. J Neurosci 2009; 29:11594–11600.

    Article  PubMed  CAS  Google Scholar 

  65. Chang PA, Wu YJ. Neuropathy target esterase: an essential enzyme for neural development and axonal maintenance. Int J Biochem Cell Biol 2010; 42:573–575.

    Article  PubMed  CAS  Google Scholar 

  66. Hargreaves AJ, Fowler, MJ, Sachana, M et al. Inhibition of neurite outgrowth in differentiating mouse N2a neuroblastoma cells by phenyl saligenin phosphate: Effects on neurofilament heavy chain phosphorylation, MAP kinase (ERK 1/2) activation and neuropathy target esterase activity. Biochemical Pharmacology 2006; 71:1240–1247.

    Article  PubMed  CAS  Google Scholar 

  67. Sachana M, Flaskos J, Alexaki E et al. The toxicity of chlorpyrifos towards differentiating mouse N2a neuroblastoma cells. Toxicol In Vitro 2001; 15:369–372.

    Article  PubMed  CAS  Google Scholar 

  68. Chang PA, Wu YJ, Chen R et al. Inhibition of neuropathy target esterase expressing by antisense RNA does not affect neural differentiation in human neuroblastoma (SK-N-SH) cell line. Mol Cell Biochem 2005; 272:47–54.

    Article  PubMed  CAS  Google Scholar 

  69. Chang PA, Chen R, Wu YJ. Reduction of neuropathy target esterase does not affect neuronal differentiation but moderate expression induces neuronal differentiation in human neuroblastoma (SK-N-SH) cell line. Mol Brain Res 2005; 141:30–38.

    Article  PubMed  CAS  Google Scholar 

  70. Chang PA, Liu CY, Chen R et al. Effect of over-expression of neuropathy target esterase on mammalian cell proliferation. Cell Prolif 2006; 39:429–440.

    Article  PubMed  Google Scholar 

  71. Suwita E, Lapadula DM, Abou-Donia MB. Calcium and calmodulin enhanced in vitro phosphorylation of hen brain cold stable microtubules and spinal cord neurofilament proteins following a single oral dose of tri-o-cresyl phosphate. Proc Natl Acad Sci USA 1986; 83:6174–6178.

    Article  PubMed  CAS  Google Scholar 

  72. Jensen KF, Lapadula DM, Knoth Anderson J et al. Anomalous phosphorylatedneurofilament aggregations in central and peripheral axons of hens treated with tri-ortho-cresyl phosphate (TOCP). J Neurosci Res 1992; 33:455–460.

    Article  PubMed  CAS  Google Scholar 

  73. Jortner BS, Perkins SK, Ehrich M. Immunohistochemical study of phosphorylated neurofilaments during the evolution of organophosphorus ester-induced delayed neuropathy. Neuro Toxicology 1999; 20:971–976.

    CAS  Google Scholar 

  74. Veeranna, Amin ND, Ahn NG et al. Mitogen-activated protein kinases (ERK 1/2) phosphorylate Lys-Ser-Pro (KSP) repeats in neurofilament proteins NFH and NFM. J Neurosci 1998; 18:4008–4021.

    PubMed  CAS  Google Scholar 

  75. Perron JC, Bixby JL. Distinct neurite outgrowth signalling pathways converge on ERK activation. Mol Cell Neurosci 1999; 13:362–378.

    Article  PubMed  CAS  Google Scholar 

  76. Sachana M, Flaskos J, Alexaki E et al. Inhibition of neurite outgrowth in N2a cells by leptophos and carbaryl: effects on neurofilament heavy chain, GAP-43, HSP-70. Toxicol In Vitro 2003; 17:115–120.

    Article  PubMed  CAS  Google Scholar 

  77. Harris W, Sachana M, Flaskos J et al. Proteomic analysis of differentiating neuroblastoma cells treated with sub-lethal neurite inhibitory concentrations of diazinon: Identification of novel biomarkers of effect. Toxicol Appl Pharmacol 2009; 240:159–165.

    Article  PubMed  CAS  Google Scholar 

  78. Carlson K, Ehrich M. Organophosphorus compounds alter F-actin content in SH-SY5Y human neuroblastoma cells. Neuro Toxicology 2001; 22:819–827.

    CAS  Google Scholar 

  79. El-Fawal HA, Ehrich MF. Calpain activity in organophosphorus-induced delayed neuropathy (OPIDN): Effects of a phenylalkylamine calcium channel blocker. Ann NY Acad Sci 1993; 679:325–329.

    Article  PubMed  CAS  Google Scholar 

  80. Song F, Yan Y, Zhao X et al. Neurofilament degradation as an early molecular event in tri-ortho-cresyl phosphate (TOCP) induced delayed neuropathy. Toxicology 2009; 258:94–100.

    Article  PubMed  CAS  Google Scholar 

  81. Flaskos J, Harris W, Sachana M et al. The effects of diazinon and cypermethrin on the differentiation of neuronal and glial cell lines. Toxicol Appl Pharmacol 2007; 219:172–180.

    Article  PubMed  CAS  Google Scholar 

  82. Gupta RP, Abou-Donia MB. Diisopropylphosphorofluoridate (DFP) treatment alters calcium activated protease activity and cytoskeletal proteins of the sciatic nerve. Brain Res 1995; 677:162–166.

    Article  PubMed  CAS  Google Scholar 

  83. Layfield R, Lowe J, Bedford L. The ubiquitin-proteasome system and neurodegenerative disorders. Essays Biochem 2005; 41:157–171.

    Article  PubMed  CAS  Google Scholar 

  84. Masoud A, Kiran R, Sandhir R. Impaired mitochondrial functions in organophosphorus induced delayed neuropathy in rats. Cell Mol Neurobiol 2009; 29:1245–1255.

    Article  PubMed  CAS  Google Scholar 

  85. Kaur P, Radotra B, Minz RZ et al. Impaired mitochondrial metabolism and neuronal apoptotic cell death after chronic dichlorvos (OP) exposure. Neuro Toxicology 2007; 28:1208–1219.

    CAS  Google Scholar 

  86. Carlson K, Ehrich M. Organophosphorus compound induced modification of SH-SY5Y human neuroblastoma mitochondrial transmembrane potential. Toxicol Appl Pharmacol 1999; 160:33–42.

    Article  PubMed  CAS  Google Scholar 

  87. Milatovic D, Gupta RC, Aschner M. Anticholinesterase activity and oxidative stress. Scientific World J 2006; 6:295–310.

    Article  CAS  Google Scholar 

  88. Mou DL, Wang YP, Song JF et al. Triorthocresyl phosphate-induced neuronal losses in lumbar spinal cord of hens—an immunohistochemistry and ultrastructure study. Int J Neurosci 2006; 116: 1303–1316.

    Article  PubMed  CAS  Google Scholar 

  89. Lockridge O, Schopfer LM. Review of tyrosine and lysine as new motifs for organophosphate binding to proteins that have no active site serine. Chem Biol Interact 2010; 187:344–388.

    Article  PubMed  CAS  Google Scholar 

  90. Li B, Schopfer LM, Hinrichs SH et al. Matrix-assisted laser desorption/ionization time of flight mass spectrometry assay for organophosphorus toxicants bound to human albumin at Tyr411. Anal Biochem 2007; 361:263–272.

    Article  PubMed  CAS  Google Scholar 

  91. Li B, Schopfer LM, Grigoryan H et al. Tyrosines of human and mouse transferrin covalently labelled by organophosphorus agents: a new motif for binding to proteins that have no active site serine. Toxicol Sci 2009; 107:144–155.

    Article  PubMed  CAS  Google Scholar 

  92. Grigoryan H, Lockridge O. Nanoimages show disruption of tubulin polymerisation by chlorpyrifosoxon: implications for neurotoxicity. Toxicol Appl Pharmacol 2009; 240:143–148.

    Article  PubMed  CAS  Google Scholar 

  93. Jiang W, Duysen EG, Hansen H et al. Mice treated with chlorpyrifos or chlorpyrifosoxon have organophosphorylated tubulin in the brain and disrupted microtubule structures, suggesting a role for tubulin in neurotoxicity associated with exposure to organophosphorus agents. Toxicol Sci 2010; 115: 183–193.

    Article  PubMed  CAS  Google Scholar 

  94. Grigoryan H, Schopfer LM, Thompson CM et al. Mass spectrometry identifies covalent binding of soman, sarin, chlorpyrifosoxon, diisopropylfluorophosphate and FP-biotin to tyrosines on tubulin: a potential mechanism of long term toxicity by organophosphorus agents. Chem Biol Interact 2008; 175: 180–186.

    Article  PubMed  CAS  Google Scholar 

  95. Grigoryan H, Schopfer LM, Peeples ES et al. Mass spectrometry identifies multiple organophosphorylated sites on tubulin. Toxicol Appl Pharmacol 2009; 240:149–158.

    Article  PubMed  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2012 Landes Bioscience and Springer Science+Business Media

About this chapter

Cite this chapter

Hargreaves, A.J. (2012). Neurodegenerations Induced by Organophosphorous Compounds. In: Ahmad, S.I. (eds) Neurodegenerative Diseases. Advances in Experimental Medicine and Biology, vol 724. Springer, New York, NY. https://doi.org/10.1007/978-1-4614-0653-2_15

Download citation

Publish with us

Policies and ethics