Skip to main content

Advertisement

Log in

Environmental neurotoxic pollutants: review

  • Review Article
  • Published:
Environmental Science and Pollution Research Aims and scope Submit manuscript

Abstract

Environmental pollutants are recognized as one of the major concerns for public health and responsible for various forms of neurological disorders. Some of the common sources of environmental pollutants related to neurotoxic manifestations are industrial waste, pesticides, automobile exhaust, laboratory waste, and burning of terrestrial waste. Among various environmental pollutants, particulate matter, ultrafine particulate matter, nanoparticles, and lipophilic vaporized toxicant (acrolein) easily cross the blood–brain barrier, activate innate immune responses in the astrocytes, microglia, and neurons, and exert neurotoxicity. Growing shreds of evidence from human epidemiological studies have correlated the environmental pollutants with neuroinflammation, oxidative stress, endoplasmic reticulum stress, mitochondrial dysfunction, myelin sheath disruption, and alterations in the blood–brain barrier anatomy leading to cognitive dysfunction and poor quality of life. These environmental pollutants also considerably cause developmental neurotoxicity, exhibit teratogenic effect and mental growth retardance, and reduce IQ level. Until now, the exact mechanism of pollutant-induced neurotoxicity is not known, but studies have shown interference of pollutants with the endogenous antioxidant defense system, inflammatory pathway (Nrf2/NF-kB, MAPKs/PI3K, and Akt/GSK3β), modulation of neurotransmitters, and reduction in long-term potentiation. In the current review, various sources of pollutants and exposure to the human population, developmental neurotoxicity, and molecular mechanism of different pollutants involved in the pathogenesis of different neurological disorders have been discussed.

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

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8
Fig. 9

Similar content being viewed by others

References

  • Aaseth J, Wallace DR, Vejrup K, Alexander J (2020) Methylmercury and developmental neurotoxicity: a global concern. Curr Opin Toxicol

  • Ailshire JA, Crimmins EM (2014) Fine particulate matter air pollution and cognitive function among older US adults. 180:359–366

  • Aitbali Y, Ba-M’hamed S, Elhidar N, Nafis A, Soraa N, Bennis M (2018) Glyphosate based-herbicide exposure affects gut microbiota, anxiety and depression-like behaviors in mice. Neurotoxicol Teratol 67:44–49

    CAS  Google Scholar 

  • Allen JL et al (2014) Developmental exposure to concentrated ambient ultrafine particulate matter air pollution in mice results in persistent and sex-dependent behavioral neurotoxicity and glial activation. Toxicol Sci 140:160–178

    CAS  Google Scholar 

  • Andersen HR, Nielsen JB, Grandjean P (2000) Toxicologic evidence of developmental neurotoxicity of environmental chemicals. 144:121–127

  • Atwood D, Paisley-Jones C (2017) Pesticides industry sales and usage: 2008–2012 Market Estimates US Environmental Protection Agency, Washington, DC 20460

  • Babadjouni RM, Hodis DM, Radwanski R, Durazo R, Patel A, Liu Q, Mack WJ (2017) Clinical effects of air pollution on the central nervous system; a review. 43:16–24

  • Bailey DC et al (2018) Chronic exposure to a glyphosate-containing pesticide leads to mitochondrial dysfunction and increased reactive oxygen species production in Caenorhabditis elegans. Environ Toxicol Pharmacol 57:46–52

    CAS  Google Scholar 

  • Bakir F et al (1973) Methylmercury poisoning in Iraq. 181:230–241

  • Bank MS, Vignati DA, Vigon B (2014) United Nations Environment Programme’s Global Mercury Partnership: science for successful implementation of the Minamata Convention. Environ Toxicol Chem 33:1199–1201

    CAS  Google Scholar 

  • Barats A, Pécheyran C, Amouroux D, Dubascoux S, Chauvaud L, Donard OFX (2007) Matrix-matched quantitative analysis of trace-elements in calcium carbonate shells by laser-ablation ICP–MS: application to the determination of daily scale profiles in scallop shell (Pecten maximus). 387:1131–1140

  • Beauchamp RO, Andjelkovich DA, Kligerman AD, Morgan KT, Heck HD (1985) A critical review of the literature on acrolein toxicity. 14:309–380

  • Bein K, Leikauf GD (2020) Acrolein and unsaturated aldehydes: 205-259

  • Belyaeva EA, Sokolova TV, Emelyanova LV, Zakharova IO (2012) Mitochondrial electron transport chain in heavy metal-induced neurotoxicity: effects of cadmium, mercury, and copper 2012

  • Bertotto LB, Catron TR, Tal TJN (2020) Exploring interactions between xenobiotics, microbiota, and neurotoxicity in zebrafish. 76:235–244

  • Bharagava RN, Saxena G, Mulla SI (2020) Introduction to industrial wastes containing organic and inorganic pollutants and bioremediation approaches for environmental management. In: Bioremediation of industrial waste for environmental safety. Springer, pp 1–18

  • Bjørklund G, Mutter J, Aaseth J (2017) Metal chelators and neurotoxicity: lead, mercury, and arsenic. 91:3787–3797

  • Brabhukumr A, Malhi P, Ravindra K, Lakshmi PVM (2020) Exposure to household air pollution during first 3 years of life and IQ level among 6–8-year-old children in India–a cross-sectional study. 709:135110

  • Bradbury M, Deane RJN (1993) Permeability of the blood-brain barrier to lead. 14:131–136

  • Bradman A, Schwartz JM, Fenster L, Barr DB, Holland NT, Eskenazi B (2007) Factors predicting organochlorine pesticide levels in pregnant Latina women living in a United States agricultural area. J Expo Sci Environ Epidemiol 17:388–399

    CAS  Google Scholar 

  • Breivik K, Sweetman A, Pacyna JM, Jones KC (2002) Towards a global historical emission inventory for selected PCB congeners—a mass balance approach: 2. Emissions. 290:199–224

  • Bridi D, Altenhofen S, Gonzalez JB, Reolon GK, Bonan CD (2017) Glyphosate and Roundup® alter morphology and behavior in zebrafish. Toxicology 392:32–39

    CAS  Google Scholar 

  • Bumbăcilă B, Putz MV (2020) Neurotoxicity of pesticides: the roadmap for the cubic mode of action. Curr Med Chem 27:54–77

    Google Scholar 

  • Burke RD (2016) A structural, functional, and behavioral evaluation of the developmental neurotoxicity of the organophosphorus insecticide chlorpyrifos in Guinea pigs: mechanistic implications

  • Burke RD, Todd SW, Lumsden E, Mullins RJ, Mamczarz J, Fawcett WP, Gullapalli RP, Randall WR, Pereira EFR, Albuquerque EX (2017) Developmental neurotoxicity of the organophosphorus insecticide chlorpyrifos: from clinical findings to preclinical models and potential mechanisms. J Neurochem 142:162–177

    CAS  Google Scholar 

  • Butenhoff JL et al (2012) Toxicological evaluation of ammonium perfluorobutyrate in rats: twenty-eight-day and ninety-day oral gavage studies. 33:513–530

  • Byers RK, Lord EE (1943) Late effects of lead poisoning on mental development. 66:471–494

  • Caito S, Aschner M (2015) Neurotoxicity of metals. In: Handbook of clinical neurology, vol 131. Elsevier, pp 169–189

  • Calderon-Garciduenas L et al (2003) DNA damage in nasal and brain tissues of canines exposed to air pollutants is associated with evidence of chronic brain inflammation and neurodegeneration. 31:524–538

  • Calderón-Garcidueñas L, Mora-Tiscareño A, Ontiveros E, Gómez-Garza G, Barragán-Mejía G, Broadway J, Chapman S, Valencia-Salazar G, Jewells V, Maronpot RR, Henríquez-Roldán C, Pérez-Guillé B, Torres-Jardón R, Herrit L, Brooks D, Osnaya-Brizuela N, Monroy ME, González-Maciel A, Reynoso-Robles R, Villarreal-Calderon R, Solt AC, Engle RW (2008) Air pollution, cognitive deficits and brain abnormalities: a pilot study with children and dogs. Brain Cogn 68:117–127

    Google Scholar 

  • Cattani D et al (2017) Developmental exposure to glyphosate-based herbicide and depressive-like behavior in adult offspring: implication of glutamate excitotoxicity and oxidative stress. Toxicology 387:67–80

    CAS  Google Scholar 

  • Ceccariglia S, Alvino A, Del Fà A, Parolini O, Michetti F, Gangitano C (2020) Autophagy is activated in vivo during trimethyltin-induced apoptotic neurodegeneration: a study in the rat hippocampus. 21:175

  • Chan MP, Morisawa S, Nakayama A, Kawamoto Y, Yoneda M (2006) Development of an in vitro blood–brain barrier model to study the effects of endosulfan on the permeability of tight junctions and a comparative study of the cytotoxic effects of endosulfan on rat and human glial and neuronal cell cultures. Environ Toxicol 21:223–235

    CAS  Google Scholar 

  • Charli A, Jin H, Anantharam V, Kanthasamy A, Kanthasamy AG (2016) Alterations in mitochondrial dynamics induced by tebufenpyrad and pyridaben in a dopaminergic neuronal cell culture model. Neurotoxicology 53:302–313

    CAS  Google Scholar 

  • Chen P, Bornhorst J, Aschner M (2019) Manganese metabolism in humans

  • Cheng SH, Wai AWK, So CH, Wu RSS (2000) Cellular and molecular basis of cadmium-induced deformities in zebrafish embryos. Environ Toxicol Chem 19:3024–3031

    CAS  Google Scholar 

  • Christen V, Rusconi M, Crettaz P, Fent K (2017) Developmental neurotoxicity of different pesticides in PC-12 cells in vitro. Toxicol Appl Pharmacol 325:25–36

    CAS  Google Scholar 

  • Costa LG, Cole TB, Coburn J, Chang Y-C, Dao K, Roqué PJ (2017) Neurotoxicity of traffic-related air pollution. 59:133–139

  • Costa LG, Cole TB, Dao K, Chang Y-C, Garrick JM (2019) Developmental impact of air pollution on brain function. 131:104580

  • Despang P, Salamon S, Breitenkamp AF, Kuzmenkina E, Herzig S, Matthes J (2020) Autism-associated mutations in the CaVβ2 calcium-channel subunit increase Ba2+-currents and lead to differential modulation by the RGK-protein Gem. 136:104721

  • Diaz-Ortiz ME, Chen-Plotkin AS (2020) Omics in neurodegenerative disease: hope or hype?

  • Domico LM, Cooper KR, Bernard LP, Zeevalk GD (2007) Reactive oxygen species generation by the ethylene-bis-dithiocarbamate (EBDC) fungicide mancozeb and its contribution to neuronal toxicity in mesencephalic cells. Neurotoxicology 28:1079–1091

    CAS  Google Scholar 

  • Duke SO (1990) Overview of herbicide mechanisms of action. Environ Health Perspect 87:263–271

    CAS  Google Scholar 

  • Edwards M, Triantafyllidou S, Best D (2009) Elevated blood lead in young children due to lead-contaminated drinking water: Washington, DC, 2001− 2004. 43:1618–1623

  • Ettinger AS, Leonard ML, Mason J (2019) CDC’s Lead Poisoning Prevention Program: a long-standing responsibility and commitment to protect children from lead exposure. 25:S5

  • Farina M, Aschner M (2017) Methylmercury-induced neurotoxicity: focus on pro-oxidative events and related consequences. In: Neurotoxicity of Metals. Springer, pp 267–286

  • Farina M, Aschner M (2019) Glutathione antioxidant system and methylmercury-induced neurotoxicity: an intriguing interplay. 1863:129285

  • Farina M, Aschner M, da Rocha JB (2017) The catecholaminergic neurotransmitter system in methylmercury-induced neurotoxicity. In: Advances in Neurotoxicology, vol 1. Elsevier, pp 47–81

  • Fenga C, Gangemi S, Alibrandi A, Costa C, Micali E (2016) Relationship between lead exposure and mild cognitive impairment. J Prev Med Hyg 57:E205

    CAS  Google Scholar 

  • Ferraz da Silva I, Freitas-Lima LC, Graceli JB, de Melo Rodrigues LC (2018) Organotins in neuronal damage, brain function, and behavior: a short review. 8:366

  • Finnegan C, Ryan D, Enright A-M, Garcia-Cabellos G (2018) A review of strategies for the detection and remediation of organotin pollution. 48:77–118

  • Fonnum F, Mariussen E (2009) Mechanisms involved in the neurotoxic effects of environmental toxicants such as polychlorinated biphenyls and brominated flame retardants. 111:1327–1347

  • Fortunati N et al (2017) Effects of environmental pollutants on signaling pathways in rat pituitary GH3 adenoma cells. 158:660–668

  • Franco R, Li S, Rodriguez-Rocha H, Burns M, Panayiotidis MI (2010) Molecular mechanisms of pesticide-induced neurotoxicity: relevance to Parkinson’s disease. 188:289–300

  • Fujimura M, Usuki F (2012) Differing effects of toxicants (methylmercury, inorganic mercury, lead, amyloid β, and rotenone) on cultured rat cerebrocortical neurons: differential expression of rho proteins associated with neurotoxicity. Toxicol Sci 126:506–514

    CAS  Google Scholar 

  • Gaballah S et al (2020) Evaluation of developmental toxicity, developmental neurotoxicity, and tissue dose in zebrafish exposed to GenX and other PFAS. Environ Health Perspect 128:047005

    Google Scholar 

  • Garabadu D, Agrawal N (2020) Naringin exhibits neuroprotection against rotenone-induced neurotoxicity in experimental rodents. NeuroMolecular Med:1–17

  • Gaum PM, Esser A, Schettgen T, Gube M, Kraus T, Lang J (2014) Prevalence and incidence rates of mental syndromes after occupational exposure to polychlorinated biphenyls. Int J Hyg Environ Health 217:765–774

    CAS  Google Scholar 

  • Gerlofs-Nijland ME et al (2010) Pulmonary and cardiovascular effects of traffic-related particulate matter: 4-week exposure of rats to roadside and diesel engine exhaust particles. Inhal Toxicol 22:1162–1173

    CAS  Google Scholar 

  • Gibson JL (2005) A plea for painted railings and painted walls of rooms as the source of lead poisoning amongst Queensland children. 120:301–304

  • Goel AD, Chowgule RV (2019) Outbreak investigation of lead neurotoxicity in children from artificial jewelry cottage industry. 24:30

  • Grandjean P, Landrigan PJ (2006) Developmental neurotoxicity of industrial chemicals. Lancet 368:2167–2178

    CAS  Google Scholar 

  • Guillette EA, Meza MM, Aquilar MG, Soto AD, Garcia IE (1998) An anthropological approach to the evaluation of preschool children exposed to pesticides in Mexico. 106:347–353

  • Gunter TE et al (2010) An analysis of the effects of Mn2+ on oxidative phosphorylation in liver, brain, and heart mitochondria using state 3 oxidation rate assays. 249:65–75

  • Guo H, Liu M, Zhang L, Wang L, Hou W, Ma Y, Ma Y (2020) The critical period for neuroprotection by estrogen replacement therapy and the potential underlying mechanisms

  • Hanna-Attisha M, LaChance J, Sadler RC, Champney Schnepp A (2016) Elevated blood lead levels in children associated with the Flint drinking water crisis: a spatial analysis of risk and public health response. 106:283–290

  • Hardingham GE, Fukunaga Y, Bading H (2002) Extrasynaptic NMDARs oppose synaptic NMDARs by triggering CREB shut-off and cell death pathways. 5:405–414

  • Harischandra DS, Ghaisas S, Zenitsky G, Jin H, Kanthasamy A, Anantharam V, Kanthasamy AG (2019) Manganese-induced neurotoxicity: new insights into protein misfolding, mitochondrial impairment and neuroinflammation. 13:654

  • Hartz AM, Bauer B, Block ML, Hong J-S, Miller DS (2008a) Diesel exhaust particles induce oxidative stress, proinflammatory signaling, and P-glycoprotein up-regulation at the blood-brain barrier The. FASEB J 22:2723–2733

    CAS  Google Scholar 

  • Hartz AM, Bauer B, Block ML, Hong J-S, Miller DS (2008b) Diesel exhaust particles induce oxidative stress, proinflammatory signaling, and P-glycoprotein up-regulation at the blood-brain barrier. 22:2723–2733

  • He X et al (2017) Developmental neurotoxicity of methamidophos in the embryo-larval stages of zebrafish. Int J Environ Res Public Health 14:23

    Google Scholar 

  • Hernandez CM et al (2015) Repeated exposure to chlorpyrifos leads to prolonged impairments of axonal transport in the living rodent brain. Neurotoxicology 47:17–26

    CAS  Google Scholar 

  • Heusinkveld HJ, Thomas GO, Lamot I, van den Berg M, Kroese AB, Westerink RH (2010) Dual actions of lindane (γ-hexachlorocyclohexane) on calcium homeostasis and exocytosis in rat PC12 cells. Toxicol Appl Pharmacol 248:12–19

    CAS  Google Scholar 

  • Ho W-C, Hsu C-C, Huang H-J, Wang H-T, Lin AM-YL (2020) Anti-inflammatory effect of AZD6244 on acrolein-induced neuroinflammation. 57:88–95

  • Hobbie KA, Rooney N, Scott RP, Anderson KA (2017) An alternative method to produce shikimic acid chemical feedstock by applying glyphosate to forage crops. Crop Sci 57:945–950

    CAS  Google Scholar 

  • Hong S, Hwang J, Kim JY, Shin KS, Kang SJ (2014) Heptachlor induced nigral dopaminergic neuronal loss and Parkinsonism-like movement deficits in mice. Exp Mol Med 46:e80–e80

    CAS  Google Scholar 

  • Hossain MM, DiCicco-Bloom E, Richardson JR (2015) Hippocampal ER stress and learning deficits following repeated pyrethroid exposure. Toxicol Sci 143:220–228

    CAS  Google Scholar 

  • Huang Y-J et al (2013a) Acrolein induces Alzheimer’s disease-like pathologies in vitro and in vivo. 217:184–191

  • Huang Y et al (2013b) Protective effects of caffeic acid and caffeic acid phenethyl ester against acrolein-induced neurotoxicity in HT22 mouse hippocampal cells. 535:146–151

  • Igarashi K, Uemura T, Kashiwagi K (2018) Acrolein toxicity at advanced age: present and future. 50:217–228

  • Iqubal A, Sharma S, Sharma K, Bhavsar A, Hussain I, Iqubal MK, Kumar R (2018) Intranasally administered pitavastatin ameliorates pentylenetetrazol-induced neuroinflammation, oxidative stress and cognitive dysfunction. 211:172–181

  • Iqubal A et al (2019a) Molecular mechanism involved in cyclophosphamide-induced cardiotoxicity: old drug with a new vision. 218:112–131

  • Iqubal A et al (2019b) Nerolidol attenuates cyclophosphamide-induced cardiac inflammation, apoptosis and fibrosis in Swiss Albino mice. 863:172666

  • Iqubal A, Sharma S, Najmi AK, Syed MA, Ali J, Alam MM, Haque SE (2019c) Nerolidol ameliorates cyclophosphamide-induced oxidative stress, neuroinflammation and cognitive dysfunction: plausible role of Nrf2 and NF-κB. 236:116867

  • Iqubal A, Syed MA, Haque MM, Najmi AK, Ali J, Haque E (2020a) Effect of nerolidol on cyclophosphamide-induced bone marrow and hematological toxicities in Swiss albino mice

  • Iqubal A, Syed MA, Najmi AK, Ali J, Haque SEJA (2020b) Ameliorative effect of nerolidol on cyclophosphamide-induced gonadal toxicity in Swiss Albino mice: biochemical-, histological-and immunohistochemical-based evidences:e13535

  • Jakaria M, Park S-Y, Haque M, Karthivashan G, Kim I-S, Ganesan P, Choi D-K (2018) Neurotoxic agent-induced injury in neurodegenerative disease model: focus on involvement of glutamate receptors. 11:307

  • Jaynes BS, Jennings J (2008) Hard surface cleaning compositions comprising certain perfluroalkyl substituted compounds. Google Patents

  • Ji H et al (2019) Associations of prenatal exposures to low levels of Polybrominated Diphenyl Ether (PBDE) with thyroid hormones in cord plasma and neurobehavioral development in children at 2 and 4 years. Environ Int 131:105010

    CAS  Google Scholar 

  • Jia Z, Misra HP (2007) Developmental exposure to pesticides zineb and/or endosulfan renders the nigrostriatal dopamine system more susceptible to these environmental chemicals later in life. Neurotoxicology 28:727–735

    CAS  Google Scholar 

  • Johansson N, Fredriksson A, Eriksson P (2008) Neonatal exposure to perfluorooctane sulfonate (PFOS) and perfluorooctanoic acid (PFOA) causes neurobehavioural defects in adult mice. Neurotoxicology 29:160–169

    CAS  Google Scholar 

  • Juberg DR, Hoberman AM, Marty S, Picut CA, Stump DG (2019) Letter to the editor regarding “safety of safety evaluation of pesticides: developmental neurotoxicity of chlorpyrifos and chlorpyrifos-methyl” by Mie et al.(environmental health. 2018. 17: 77). 18:21

  • Julvez J, Grandjean P (2009) Neurodevelopmental toxicity risks due to occupational exposure to industrial chemicals during pregnancy. 47:459–468

  • Jursa T, Smith DR (2009) Ceruloplasmin alters the tissue disposition and neurotoxicity of manganese, but not its loading onto transferrin. 107:182–193

  • Ke T, Sidoryk-Wegrzynowicz M, Pajarillo E, Rizor A, Soares FAA, Lee E, Aschner MJNR (2019) Role of astrocytes in manganese neurotoxicity revisited:1-11

  • Khosya S, Kushwaha S, Dung AAD (2018) Polyvinyl chloride (PVC) acting as occupational hazard in a factory worker presented with acute toxic encephalopathy: a case report. 8:2161–0495.1000400

  • Kim KH et al (2011) Para-and ortho-substitutions are key determinants of polybrominated diphenyl ether activity toward ryanodine receptors and neurotoxicity. 119:519–526

  • Kim K-S, Lee Y-M, Lee H-W, Jacobs DR Jr, Lee D-H (2015) Associations between organochlorine pesticides and cognition in US elders: National Health and Nutrition Examination Survey 1999–2002. Environ Int 75:87–92

    CAS  Google Scholar 

  • King ED, Mahoney J, Krajewski D, Smith W, Mathias C, Bose DD (2019) Effects of brominated flame retardants on cytokine production by mast cells. 33:505.502–505.502

  • Kitaguchi Y, Taraseviciene-Stewart L, Hanaoka M, Natarajan R, Kraskauskas D, Voelkel NF (2012) Acrolein induces endoplasmic reticulum stress and causes airspace enlargement 7

  • Kitazawa M, Anantharam V, Kanthasamy AG (2001) Dieldrin-induced oxidative stress and neurochemical changes contribute to apoptopic cell death in dopaminergic cells. Free Radic Biol Med 31:1473–1485

    CAS  Google Scholar 

  • Klocke C, Lein PJ (2020) Evidence implicating non-dioxin-like congeners as the key mediators of polychlorinated biphenyl (PCB) developmental neurotoxicity. Int J Mol Sci 21:1013

    CAS  Google Scholar 

  • Kochmanski J, VanOeveren SE, Patterson JR, Bernstein AI (2019) Developmental dieldrin exposure alters DNA methylation at genes related to dopaminergic neuron development and Parkinson’s disease in mouse midbrain. Toxicol Sci 169:593–607

    CAS  Google Scholar 

  • Kodavanti PRS et al (1998) Repeated exposure of adult rats to Aroclor 1254 causes brain region-specific changes in intracellular Ca2+ buffering and protein kinase C activity in the absence of changes in tyrosine hydroxylase. 153:186–198

  • Korbas M, Blechinger SR, Krone PH, Pickering IJ, George GN (2008) Localizing organomercury uptake and accumulation in zebrafish larvae at the tissue and cellular level. Proc Natl Acad Sci 105:12108–12112

    CAS  Google Scholar 

  • Kudo N (2015) Metabolism and pharmacokinetics. Intoxicological effects of perfluoroalkyl and polyfluoroalkyl substances (J. C. Jamie Ed). Humana Press (Springer International Publishing AG Switzerland), Cham…

  • Landrigan P, Baloh R, Barthel W, Whitworth R, Staehling N, Rosenblum BJTL (1975) Neuropsychological dysfunction in children with chronic low-level lead absorption. 305:708–712

  • Lange J, Condello III (2017) Lead: an environmental neurotoxic agent. 2:13

  • Lau C et al (2003) Exposure to perfluorooctane sulfonate during pregnancy in rat and mouse. II: postnatal evaluation. 74:382–392

  • Lau C, Anitole K, Hodes C, Lai D, Pfahles-Hutchens A, Seed J (2007) Perfluoroalkyl acids: a review of monitoring and toxicological findings. 99:366–394

  • Lee S et al (2016) Trimethyltin-induced hippocampal neurodegeneration: a mechanism-based review. 125:187–199

  • Lei L et al (2018) Polystyrene (nano) microplastics cause size-dependent neurotoxicity, oxidative damage and other adverse effects in Caenorhabditis elegans. Environ Sci Nano 5:2009–2020

    CAS  Google Scholar 

  • Li P, Li Z-H (2020) Environmental co-exposure to TBT and Cd caused neurotoxicity and thyroid endocrine disruption in zebrafish, a three-generation study in a simulated environment. 259:113868

  • Li Z et al (2017) Evaluation of PFOS-mediated neurotoxicity in rat primary neurons and astrocytes cultured separately or in co-culture. 38:77–90

  • Li H et al (2019a) Role of Nrf2 in the antioxidation and oxidative stress induced developmental toxicity of honokiol in zebrafish. 373:48–61

  • Li Y, Lin N, Ji X, Mai J, Li Q (2019b) Organotin compound DBDCT induces CYP3A suppression through NF-κB-mediated repression of PXR activity. Metallomics 11:936–948

    CAS  Google Scholar 

  • Li J et al (2020) Lactational exposure of polybrominated diphenyl ethers and its association with infant developmental measurements. J Hazard Mater:122031

  • Liang G, Qin H, Zhang L, Ma S, Huang X, Lv Y, Qing L, Li Q, Xiong Y, Huang Y, Chen K, Huang Y, Shen Y, Nong J, Yang X, Zou Y (2015) Effects of chronic manganese exposure on the learning and memory of rats by observing the changes in the hippocampal cAMP signaling pathway. Food Chem Toxicol 83:261–267

    CAS  Google Scholar 

  • Liu J et al (2019) A protective role of autophagy in Pb-induced developmental neurotoxicity in zebrafish. Chemosphere 235:1050–1058

    CAS  Google Scholar 

  • Liu J-H et al (2020) Acrolein is involved in ischemic stroke-induced neurotoxicity through spermidine/spermine-N1-acetyltransferase activation. 323:113066

  • Long J et al (2019) NLRP3 inflammasome activation is involved in trimethyltin-induced neuroinflammation. 1718:186–193

  • Lovell MA, Xie C, Markesbery WR, Medicine (2000) Acrolein, a product of lipid peroxidation, inhibits glucose and glutamate uptake in primary neuronal cultures 29:714-720

  • Lubick N (2010) Endosulfan’s exit: US EPA pesticide review leads to a ban. American Association for the Advancement of Science

  • Luebker DJ, Case MT, York RG, Moore JA, Hansen KJ, Butenhoff JL (2005) Two-generation reproduction and cross-foster studies of perfluorooctanesulfonate (PFOS) in rats. 215:126–148

  • Mao G et al (2020) Evaluation of combined developmental neurological toxicity of di (n-butyl) phthalates and lead using immature mice. Environ Sci Pollut Res:1–9

  • Mariussen E (2012) Neurotoxic effects of perfluoroalkylated compounds: mechanisms of action and environmental relevance. 86:1349–1367

  • Medda N, Patra R, Ghosh TK, Maiti S (2020) Neurotoxic mechanism of arsenic: synergistic effect of mitochondrial instability, oxidative stress, and hormonal-neurotransmitter impairment:1-8

  • Mishra S, Bharagava RN, More N, Yadav A, Zainith S, Mani S, Chowdhary P (2019) Heavy metal contamination: an alarming threat to environment and human health. In: Environmental biotechnology: for sustainable future. Springer, pp 103–125

  • Mitra S, Gera R, Siddiqui WA, Khandelwal S (2013) Tributyltin induces oxidative damage, inflammation and apoptosis via disturbance in blood–brain barrier and metal homeostasis in cerebral cortex of rat brain: an in vivo and in vitro study. Toxicology 310:39–52

    CAS  Google Scholar 

  • Morris-Schaffer K, Merrill AK, Wong C, Jew K, Sobolewski M, Cory-Slechta DA (2019) Limited developmental neurotoxicity from neonatal inhalation exposure to diesel exhaust particles in C57BL/6 mice. 16:1–14

  • Mortuza TB, Edwards GL, White CA, Patel V, Cummings BS, Bruckner JV (2019) Age dependency of blood-brain barrier penetration by cis-and trans-permethrin in the rat. 47:234–237

  • Moser V, McGee J, Ehman KD (2008) Concentration and persistence of tin in rat brain and blood following dibutyltin exposure during development. 72:47–52

  • Mursaleen L, Somavarapu S, Zariwala MG (2019) Deferoxamine and curcumin loaded nanocarriers protect against rotenone-induced neurotoxicity. J Parkinsons Dis:1–13

  • Neal AP, Stansfield KH, Worley PF, Thompson RE, Guilarte TR (2010) Lead exposure during synaptogenesis alters vesicular proteins and impairs vesicular release: potential role of NMDA receptor–dependent BDNF signaling. 116:249–263

  • Nedzvetskii V, Kirichenko S, Baydas G, Nerush OP (2012) Effects of melatonin on memory and learning deficits induced by exposure to thinner. 44:42–48

  • OECD (2018) Toward a New Comprehensive Global Database of Per- And Polyfluoroalkyl Substances (PFASs): Summary Report on Updating the OECD 2007 List of Per- and Polyfluoroalkyl Substances (PFASs)”. Paris : OECD., ENV/JM/MONO(2018)7

  • Omura M et al (2004) Distribution of tributyltin, dibutyltin and monobutyltin in the liver, brain and fat of rats: two-generation toxicity study of tributyltin chloride. 11:123–132

  • Oudin A (2020) Short review: air pollution, noise and lack of greenness as risk factors for Alzheimer’s disease-epidemiologic and experimental evidence. 134:104646

  • Park E, Chun HS (2016) Protective effects of quercetin on dieldrin-induced endoplasmic reticulum stress and apoptosis in dopaminergic neuronal cells. Neuroreport 27:1140–1146

    CAS  Google Scholar 

  • Petronilho F, Goldman JL, Barichello T (2019) Evans blue-albumin as a marker to evaluate blood-brain barrier integrity in neonatal and adult rodents. In: Blood-brain barrier. Springer, pp 197–203

  • Putra M, Gage M, Sharma S, Gardner C, Gasser G, Anantharam V, Thippeswamy T (2020) Diapocynin, an NADPH oxidase inhibitor, counteracts diisopropylfluorophosphate-induced long-term neurotoxicity in the rat model

  • Qiao D, Seidler FJ, Slotkin TA (2005) Oxidative mechanisms contributing to the developmental neurotoxicity of nicotine and chlorpyrifos Toxicology and applied pharmacology. 206:17–26

  • Richardson JR, Fitsanakis V, Westerink RH, Kanthasamy AG (2019) Neurotoxicity of pesticides. Acta Neuropathol:1–20

  • Rocha A, Trujillo KJN (2019) Neurotoxicity of low-level lead exposure: history, mechanisms of action, and behavioral effects in humans and preclinical models

  • Roda E, Coccini T, Acerbi D, Castoldi A, Bernocchi G, Manzo L (2008) Cerebellum cholinergic muscarinic receptor (subtype-2 and-3) and cytoarchitecture after developmental exposure to methylmercury: an immunohistochemical study in rat. 35:285–294

  • Rosenzweig MR, Villagran RJS (2020) Is fish brain food or brain poison? Sea surface temperature, methyl-mercury and child cognitive development. National Bureau of Economic Research

  • Saitoh M et al (2001) Tributyltin or triphenyltin inhibits aromatase activity in the human granulosa-like tumor cell line KGN. 289:198–204

  • Salvi A, Salim S (2019) Neurobehavioral consequences of traffic-related air pollution. 13:1232

  • Salvi A, Liu H, Salim S (2020) Involvement of oxidative stress and mitochondrial mechanisms in air pollution-related neurobiological impairments. 12:100205

  • Sarkar S et al (2018) Manganese exposure induces neuroinflammation by impairing mitochondrial dynamics in astrocytes. 64:204–218

  • Schaafsma SM, Pfaff DW (2014) Etiologies underlying sex differences in autism spectrum disorders. Front Neuroendocrinol 35:255–271

    Google Scholar 

  • Schirinzi GF, Pérez-Pomeda I, Sanchís J, Rossini C, Farré M, Barceló D (2017) Cytotoxic effects of commonly used nanomaterials and microplastics on cerebral and epithelial human cells. Environ Res 159:579–587

    CAS  Google Scholar 

  • Schmidt JT, Rushin A, Boyda J, Souders CL II, Martyniuk CJ (2017) Dieldrin-induced neurotoxicity involves impaired mitochondrial bioenergetics and an endoplasmic reticulum stress response in rat dopaminergic cells. Neurotoxicology 63:1–12

    CAS  Google Scholar 

  • Seaton A, Tran L, Chen R, Maynard RL, Whalley LJ (2020) Pollution, particles, and dementia: a hypothetical causative pathway. 17:862

  • Sen S, Flynn MR, Du G, Tröster AI, An H, Huang XJTS (2011) Manganese accumulation in the olfactory bulbs and other brain regions of “asymptomatic” welders. 121:160–167

  • Shafer TJ, Meyer DA, Crofton KM (2005) Developmental neurotoxicity of pyrethroid insecticides: critical review and future research needs. Environ Health Perspect 113:123–136

    CAS  Google Scholar 

  • Sharma H, Zhang P, Barber DS, Liu B (2010) Organochlorine pesticides dieldrin and lindane induce cooperative toxicity in dopaminergic neurons: role of oxidative stress. Neurotoxicology 31:215–222

    CAS  Google Scholar 

  • Shi R, Page J, Tully M (2015) Molecular mechanisms of acrolein-mediated myelin destruction in CNS trauma and disease. 49:888–895

  • Shrot S, Tauber M, Shiyovich A, Milk N, Rosman Y, Eisenkraft A, Kadar T, Kassirer M, Cohen Y (2015) Early brain magnetic resonance imaging can predict short and long-term outcomes after organophosphate poisoning in a rat model. Neurotoxicology 48:206–216

    CAS  Google Scholar 

  • Silva MH, Gammon D (2009) An assessment of the developmental, reproductive, and neurotoxicity of endosulfan Birth defects research part b: developmental and reproductive. Toxicology 86:1–28

    CAS  Google Scholar 

  • Slotkin TA, Skavicus S, Stapleton HM, Seidler FJJT (2017) Brominated and organophosphate flame retardants target different neurodevelopmental stages, characterized with embryonic neural stem cells and neuronotypic PC12 cells. 390:32–42

  • Soderlund DM (2012) Molecular mechanisms of pyrethroid insecticide neurotoxicity: recent advances. Arch Toxicol 86:165–181

    CAS  Google Scholar 

  • Spratlen MJ et al (2020) The association between prenatal exposure to perfluoroalkyl substances and childhood neurodevelopment. Environ Pollut:114444

  • Sukharenko E, Samoylova I, Nedzvetsky V (2017) Molecular mechanisms of aluminium ions neurotoxicity in brain cells of fish from various pelagic areas Regulatory Mechanisms in Biosystems 3

  • Sun R, Gu DA (2008) Air pollution, economic development of communities, and health status among the elderly in urban China. 168:1311–1318

  • Tanner CM, Kamel F, Ross GW, Hoppin JA, Goldman SM, Korell M, Marras C, Bhudhikanok GS, Kasten M, Chade AR, Comyns K, Richards MB, Meng C, Priestley B, Fernandez HH, Cambi F, Umbach DM, Blair A, Sandler DP, Langston JW (2011) Rotenone, paraquat, and Parkinson’s disease. Environ Health Perspect 119:866–872

    CAS  Google Scholar 

  • Todt CE et al (2016) Acute exposure to a Mn/Zn ethylene-bis-dithiocarbamate fungicide leads to mitochondrial dysfunction and increased reactive oxygen species production in Caenorhabditis elegans. Neurotoxicology 57:112–120

    CAS  Google Scholar 

  • Toesca A, Geloso MC, Mongiovì AM, Furno A, Schiattarella A, Michetti F, Corvino V (2016) Trimethyltin modulates reelin expression and endogenous neurogenesis in the hippocampus of developing rats. Neurochem Res 41:1559–1569

    CAS  Google Scholar 

  • Toxicant D (2019) PFOA and PFOS

  • Trumbo P, Yates AA, Schlicker S, Poos M (2001) Dietary reference intakes: vitamin A, vitamin K, arsenic, boron, chromium, copper, iodine, iron, manganese, molybdenum, nickel, silicon, vanadium, and zinc. 101:294

  • Tukker AM, Wijnolts FM, de Groot A, Westerink RH (2018) Human iPSC-derived neuronal models for in vitro neurotoxicity assessment. Neurotoxicology 67:215–225

    CAS  Google Scholar 

  • Vargas R, Ponce-Canchihuamán J (2017) Emerging various environmental threats to brain and overview of surveillance system with zebrafish model. 4:467–473

  • Vickers NJ (2017) Animal communication: when I’m calling you, will you answer too? 27:R713–R715

  • Wang Q, Luo W, Zhang W, Liu M, Song H, Chen J (2011) Involvement of DMT1+ IRE in the transport of lead in an in vitro BBB model. 25:991–998

  • Wang Y, Wang L, Chang W, Zhang Y, Zhang Y, Liu W (2019) Neurotoxic effects of perfluoroalkyl acids: neurobehavioral deficit and its molecular mechanism. 305:65–72

  • Weiss B (2000) Vulnerability of children and the developing brain to neurotoxic hazards. 108:375–381

  • Weiss JM, Andersson PL, Lamoree MH, Leonards PE, van Leeuwen SP, Hamers T (2009) Competitive binding of poly-and perfluorinated compounds to the thyroid hormone transport protein transthyretin. 109:206–216

  • Williams TI, Lynn BC, Markesbery WR, Lovell MA (2006) Increased levels of 4-hydroxynonenal and acrolein, neurotoxic markers of lipid peroxidation, in the brain in mild cognitive impairment and early Alzheimer’s disease. 27:1094–1099

  • Wolfe M, McDonald N, Arunachalam S, Valencia A (2017) Air pollution exposure during school commutes. 5:S48–S49

  • Xiao C, Gu Y, Zhou C-Y, Wang L, Zhang M-M, Ruan D-Y (2006) Pb2+ impairs GABAergic synaptic transmission in rat hippocampal slices: a possible involvement of presynaptic calcium channels. 1088:93–100

  • Xu J et al (2019) Mercury methylating microbial communities of boreal forest soils. 9:1–13

  • Yamashita N, Doi M, Nishio M, Hojo H, Tanaka M (1972) Recent observations of Kyoto children poisoned by arsenic tainted “Morinaga dry milk” Nippon Eiseigaku Zasshi (Japanese Journal of Hygiene) 27:364-399

  • Yan R, Page JC, Shi R (2016) Acrolein-mediated conduction loss is partially restored by K+ channel blockers. 115:701–710

  • Yang T, Xu Z, Liu W, Xu B, Deng YJET (2020) Oxidative stress accelerates synaptic glutamate dyshomeostasis and NMDARs disorder during methylmercury-induced neuronal apoptosis in rat cerebral cortex

  • Yuchi W, Sbihi H, Davies H, Tamburic L, Brauer MJEH (2020) Road proximity, air pollution, noise, green space and neurologic disease incidence: a population-based cohort study 19:8

  • Zarazúa S, Bürger S, Delgado JM, Jiménez-Capdeville ME, Schliebs R (2011) Arsenic affects expression and processing of amyloid precursor protein (APP) in primary neuronal cells overexpressing the Swedish mutation of human APP. Int J Dev Neurosci 29:389–396

    Google Scholar 

  • Zeng HC et al (2011) Prenatal exposure to perfluorooctanesulfonate in rat resulted in long-lasting changes of expression of synapsins and synaptophysin. 65:225–233

  • Zeng R et al (2019) Icariin-mediated activation of autophagy confers protective effect on rotenone induced neurotoxicity in vivo and in vitro. Toxicol Rep 6:637–644

    CAS  Google Scholar 

  • Zhang S, Ireland D, Sipes NS, Behl M, Collins EMS (2019) Screening for neurotoxic potential of 15 flame retardants using freshwater planarians. 73:54–66

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Syed Ehtaishamul Haque.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Additional information

Responsible editor: Mohamed M. Abdel-Daim

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Iqubal, A., Ahmed, M., Ahmad, S. et al. Environmental neurotoxic pollutants: review. Environ Sci Pollut Res 27, 41175–41198 (2020). https://doi.org/10.1007/s11356-020-10539-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11356-020-10539-z

Keywords

Navigation