1932

Abstract

Brain development is impaired by maternal exposure to airborne toxins from ambient air pollution, cigarette smoke, and lead. Shared postnatal consequences include gray matter deficits and abnormal behaviors as well as elevated blood pressure. These unexpectedly broad convergences have implications for later life brain health because these same airborne toxins accelerate brain aging. Gene-environment interactions are shown for alleles that influence the risk of Alzheimer disease. The multigenerational trace of these toxins extends before fertilization because egg cells are formed in the grandmaternal uterus. The lineage and sex-specific effects of grandmaternal exposure to lead and cigarettes indicate epigenetic processes of relevance to future generations from our current and recent exposure to airborne toxins.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-devpsych-042320-044338
2020-12-15
2024-05-02
Loading full text...

Full text loading...

/deliver/fulltext/devpsych/2/1/annurev-devpsych-042320-044338.html?itemId=/content/journals/10.1146/annurev-devpsych-042320-044338&mimeType=html&fmt=ahah

Literature Cited

  1. Abraham M, Alramadhan S, Iniguez C, Duijts L, Jaddoe VW et al. 2017. A systematic review of maternal smoking during pregnancy and fetal measurements with meta-analysis. PLOS ONE 12:e0170946
    [Google Scholar]
  2. Agency Toxic Subst. Dis. Registry 2019. Toxicological profile for lead: draft for the public comment, May 2019 Rep., Agency Toxic Subst. Dis. Registry, Atlanta https://www.atsdr.cdc.gov/toxprofiles/tp13.pdf
    [Google Scholar]
  3. Ailshire JA, Crimmins EM. 2014. Fine particulate matter air pollution and cognitive function among older US adults. Am. J. Epidemiol. 180:359–66
    [Google Scholar]
  4. Allen JL, Liu X, Pelkowski S, Palmer B, Conrad K et al. 2014a. Early postnatal exposure to ultrafine particulate matter air pollution: persistent ventriculomegaly, neurochemical disruption, and glial activation preferentially in male mice. Environ. Health Perspect. 122:939–45
    [Google Scholar]
  5. Allen JL, Liu X, Weston D, Prince L, Oberdörster G et al. 2014b. 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. 140:160–78
    [Google Scholar]
  6. Allen JL, Oberdorster G, Morris-Schaffer K, Wong C, Klocke C et al. 2017. Developmental neurotoxicity of inhaled ambient ultrafine particle air pollution: parallels with neuropathological and behavioral features of autism and other neurodevelopmental disorders. Neurotoxicology 59:140–54
    [Google Scholar]
  7. Amos-Kroohs RM, Williams MT, Braun AA, Graham DL, Webb CL et al. 2013. Neurobehavioral phenotype of C57BL/6J mice prenatally and neonatally exposed to cigarette smoke. Neurotoxicol. Teratol. 35:34–45
    [Google Scholar]
  8. Apata M, Pfeifer SP. 2020. Recent population genomic insights into the genetic basis of arsenic tolerance in humans: the difficulties of identifying positively selected loci in strongly bottlenecked populations. Heredity 124:253–62
    [Google Scholar]
  9. Arrifano GPF, Martín-Doimeadios RCR, Jiménez-Moreno M, Fernández-Trujillo S, Augusto-Oliveira M et al. 2018. Genetic susceptibility to neurodegeneration in Amazon: apolipoprotein E genotyping in vulnerable populations exposed to mercury. Front. Genet. 9:285
    [Google Scholar]
  10. Ashraf MW. 2012. Levels of heavy metals in popular cigarette brands and exposure to these metals via smoking. Sci. World J. 2012:729430
    [Google Scholar]
  11. Beckwith T, Cecil K, Altaye M, Severs R, Wolfe C et al. 2020. Reduced gray matter volume and cortical thickness associated with traffic-related air pollution in a longitudinally studied pediatric cohort. PLOS ONE 15:1e0228092
    [Google Scholar]
  12. Bernard SM, McGeehin MA. 2003. Prevalence of blood lead levels >5 μg/dL among US children 1 to 5 years of age and socioeconomic and demographic factors associated with blood of lead levels 5 to 10 μg/dL, Third National Health and Nutrition Examination Survey, 1988–1994. Pediatrics 112:1308–13
    [Google Scholar]
  13. Bihaqi SW, Bahmani A, Adem A, Zawia NH 2014. Infantile postnatal exposure to lead (Pb) enhances tau expression in the cerebral cortex of aged mice: relevance to AD. Neurotoxicology 44:114–20
    [Google Scholar]
  14. Bolton JL, Auten RL, Bilbo SD 2014. Prenatal air pollution exposure induces sexually dimorphic fetal programming of metabolic and neuroinflammatory outcomes in adult offspring. Brain Behav. Immun. 37:30–44
    [Google Scholar]
  15. Bolton JL, Huff NC, Smith SH, Mason SN, Foster WM et al. 2013. Maternal stress and effects of prenatal air pollution on offspring mental health outcomes in mice. Environ. Health Perspect. 121:1075–82
    [Google Scholar]
  16. Bolton JL, Marinero S, Hassanzadeh T, Natesan D, Le D et al. 2017. Gestational exposure to air pollution alters cortical volume, microglial morphology, and microglia-neuron interactions in a sex-specific manner. Front. Synaptic Neurosci. 9:10
    [Google Scholar]
  17. Bolton JL, Smith SH, Huff NC, Gilmour MI, Foster WM et al. 2012. Prenatal air pollution exposure induces neuroinflammation and predisposes offspring to weight gain in adulthood in a sex-specific manner. FASEB J 26:4743–54
    [Google Scholar]
  18. Braak H, Thal DR, Ghebremedhin E, Del Tredici K 2011. Stages of the pathologic process in Alzheimer disease: age categories from 1 to 100 years. J. Neuropathol. Exp. Neurol. 70:11960–69
    [Google Scholar]
  19. Breton CV, Mack WJ, Yao J, Berhane K, Amadeus M et al. 2016. Prenatal air pollution exposure and early cardiovascular phenotypes in young adults. PLOS ONE 11:3e0150825
    [Google Scholar]
  20. Buck JM, O'Neill HC, Stitzel JA 2020. Developmental nicotine exposure engenders intergenerational downregulation and aberrant posttranslational modification of cardinal epigenetic factors in the frontal cortices, striata, and hippocampi of adolescent mice. Epigenetics Chromatin 13:13
    [Google Scholar]
  21. Buck JM, Sanders KN, Wageman CR, Knopik VS, Stitzel JA, O'Neill HC 2019. Developmental nicotine exposure precipitates multigenerational maternal transmission of nicotine preference and ADHD-like behavioral, rhythmometric, neuropharmacological, and epigenetic anomalies in adolescent mice. Neuropharmacology 149:66–82
    [Google Scholar]
  22. Budtz-Jørgensen E, Bellinger D, Lanphear B, Grandjean PInt. Pool. Lead Study Investig 2013. An international pooled analysis for obtaining a benchmark dose for environmental lead exposure in children. Risk Anal. 33:450–61
    [Google Scholar]
  23. Burnett R, Chen H, Szyszkowicz M, Fann N, Hubbell B et al. 2018. Global estimates of mortality associated with long-term exposure to outdoor fine particulate matter. PNAS 115:9592–97
    [Google Scholar]
  24. Cacciottolo M, Morgan TE, Saffari AA, Shirmohammadi F, Forman HJ et al. 2020. Traffic-related air pollutants (TRAP-PM) promote neuronal amyloidogenesis through oxidative damage to lipid rafts. Free Radic. Biol. Med. 147:242–51
    [Google Scholar]
  25. Cacciottolo M, Wang X, Driscoll I, Woodward N, Saffari A et al. 2017. Particulate air pollutants, APOE alleles and their contributions to cognitive impairment in older women and to amyloidogenesis in experimental models. Transl. Psychiatry 7:e1022
    [Google Scholar]
  26. Calderón-Garcidueñas L, Herrera-Soto A, Jury N, Maher BA, González-Maciel A et al. 2020a. Reduced repressive epigenetic marks, increased DNA damage and Alzheimer's disease hallmarks in the brain of humans and mice exposed to particulate urban air pollution. Environ. Res. 183:109226
    [Google Scholar]
  27. Calderón-Garcidueñas L, Torres-Jardón R, Kulesza RJ, Mansour Y, González-González LO et al. 2020b. Alzheimer disease starts in childhood in polluted Metropolitan Mexico City. A major health crisis in progress. Environ. Res. 183:109137
    [Google Scholar]
  28. Carmines EL, Rajendran N. 2008. Evidence for carbon monoxide as the major factor contributing to lower fetal weights in rats exposed to cigarette smoke. Toxicol. Sci. 102:383–91
    [Google Scholar]
  29. Cecil KM, Brubaker CJ, Adler CM, Dietrich KN, Altaye M et al. 2008. Decreased brain volume in adults with childhood lead exposure. PLOS Med 5:e112
    [Google Scholar]
  30. Chan YL, Saad S, Pollock C, Oliver B, Al-Odat I et al. 2016. Impact of maternal cigarette smoke exposure on brain inflammation and oxidative stress in male mice offspring. Sci. Rep. 6:25881
    [Google Scholar]
  31. Chen M, Wang X, Hu Z, Zhou H, Xu Y et al. 2017. Programming of mouse obesity by maternal exposure to concentrated ambient fine particles. Part. Fibre Toxicol. 14:20
    [Google Scholar]
  32. Cheng L, Zhang B, Huo W, Cao Z, Liu W et al. 2017. Fetal exposure to lead during pregnancy and the risk of preterm and early-term deliveries. Int. J. Hyg. Environ. Health 220:984–89
    [Google Scholar]
  33. Cory-Slechta D, Sobolewski M, Varma G, Schneider JS 2017. Developmental lead and/or prenatal stress exposures followed by different types of behavioral experience result in the divergence of brain epigenetic profiles in a sex, brain region, and time-dependent manner: implications for neurotoxicology. Curr. Opin. Toxicol. 6:60–70
    [Google Scholar]
  34. Cory-Slechta DA, Sobolewski M, Marvin E, Conrad K, Merrill A et al. 2019. The impact of inhaled ambient ultrafine particulate matter on developing brain: potential importance of elemental contaminants. Toxicol. Pathol. 47:976–92
    [Google Scholar]
  35. Costa LG, Cole TB, Dao K, Chang YC, Coburn J, Garrick JM 2020. Effects of air pollution on the nervous system and its possible role in neurodevelopmental and neurodegenerative disorders. Pharmacol. Ther. 9:107523
    [Google Scholar]
  36. Costa LG, Cole TB, Dao K, Chang YC, Garrick JM 2019. Developmental impact of air pollution on brain function. Neurochem. Int. 131:104580
    [Google Scholar]
  37. Davis DA, Bortolato M, Godar SC, Sander TK, Iwata N et al. 2013. Prenatal exposure to urban air nanoparticles in mice causes altered neuronal differentiation and depression-like responses. PLOS ONE 8:e64128
    [Google Scholar]
  38. Deal JA, Power MC, Palta P, Alonso A, Schneider ALC et al. 2020. Relationship of cigarette smoking and time of quitting with incident dementia and cognitive decline. J. Am. Geriatr. Soc. 68:337–45
    [Google Scholar]
  39. Diamanti A, Papadakis S, Schoretsaniti S, Rovina N, Vivilaki V et al. 2019. Smoking cessation in pregnancy: an update for maternity care practitioners. Tob. Induc. Dis. 17:57
    [Google Scholar]
  40. Dignam T, Pomales A, Werner L, Newbern EC, Hodge J et al. 2019. Assessment of child lead exposure in a Philadelphia community, 2014. J. Public Health Manag. Pract. 25:53–61
    [Google Scholar]
  41. Dix-Cooper L, Eskenazi B, Romero C, Balmes J, Smith KR 2012. Neurodevelopmental performance among school age children in rural Guatemala is associated with prenatal and postnatal exposure to carbon monoxide, a marker for exposure to woodsmoke. Neurotoxicology 33:246–54
    [Google Scholar]
  42. Drake P, Driscoll AK, Mathews TJ 2018. Cigarette smoking during pregnancy: United States, 2016 Data Brief 305, Nat. Cent. Health Stat., Hyattsville, MD
  43. Dou JF, Farooqui Z, Faulk CD, Barks AK, Jones T et al. 2019. Perinatal lead (Pb) exposure and cortical neuron-specific DNA methylation in male mice. Genes 10:274
    [Google Scholar]
  44. Du Y, Ge MM, Xue W, Yang QQ, Wang S et al. 2015. Chronic lead exposure and mixed factors of gender×age×brain regions interactions on dendrite growth, spine maturity and NDR kinase. PLOS ONE 10:e0138112
    [Google Scholar]
  45. Durazzo TC, Mattsson N, Weiner MWAlzheimer's Dis. Neuroimaging Initiat 2014. Smoking and increased Alzheimer's disease risk: a review of potential mechanisms. Alzheimer's Dement 10:Suppl. 3S122–45
    [Google Scholar]
  46. Ehsanifar M, Jafari AJ, Nikzad H, Zavareh MS, Atlasi MA et al. 2019. Prenatal exposure to diesel exhaust particles causes anxiety, spatial memory disorders with alters expression of hippocampal pro-inflammatory cytokines and NMDA receptor subunits in adult male mice offspring. Ecotoxicol. Environ. Saf. 176:34–41
    [Google Scholar]
  47. Eid A, Bihaqi SW, Hemme C, Gaspar JM, Hart RP, Zawia NH 2018. Histone acetylation maps in aged mice developmentally exposed to lead: epigenetic drift and Alzheimer-related genes. Epigenomics 10:573–83
    [Google Scholar]
  48. Eid A, Bihaqi SW, Renehan WE, Zawia NH 2016. Developmental lead exposure and lifespan alterations in epigenetic regulators and their correspondence to biomarkers of Alzheimer's disease. Alzheimer's Dement 2:123–31
    [Google Scholar]
  49. Ekblad M, Korkeila J, Parkkola R, Lapinleimu H, Haataja L et al.(PIPARI Study Group). PIPARI Study Group 2010. Maternal smoking during pregnancy and regional brain volumes in preterm infants. J. Pediatr. 156:185–90
    [Google Scholar]
  50. Elten M, Donelle J, Lima I, Burnett RT, Weichenthal S et al. 2020. Ambient air pollution and incidence of early-onset paediatric type 1 diabetes: a retrospective population-based cohort study. Environ. Res. 184:109291
    [Google Scholar]
  51. Engstrom AK, Snyder JM, Maeda N, Xia Z 2017. Gene-environment interaction between lead and Apolipoprotein E4 causes cognitive behavior deficits in mice. Mol. Neurodegener. 12:114
    [Google Scholar]
  52. EPA (US Environ. Prot. Agency) 1996. EPA takes final step in phaseout of leaded gasoline Press release, Jan. 29. https://archive.epa.gov/epa/aboutepa/epa-takes-final-step-phaseout-leaded-gasoline.html
  53. Farzan SF, Howe CG, Chen Y, Gilbert-Diamond D, Cottingham KL et al. 2018. Prenatal lead exposure and elevated blood pressure in children. Environ. Int. 121:Part 21289–96
    [Google Scholar]
  54. Finch CE. 2018. Global Air Pollution in Aging and Disease: Reading Smoke Signals San Diego, CA: Academic
  55. Finch CE, Austad SN. 2012. Primate aging in the mammalian scheme: the puzzle of extreme variation in brain aging. Age 34:1075–91
    [Google Scholar]
  56. Finch CE, Kulminski AM. 2019. The Alzheimer's disease exposome. Alzheimer's Dement 15:1123–32
    [Google Scholar]
  57. Finch CE, Loehlin JC. 1998. Environmental influences that may precede fertilization: a first examination of the prezygotic hypothesis from maternal age influences on twins. Behav. Genet. 28:101–6
    [Google Scholar]
  58. Forman HJ, Finch CE. 2018. A critical review of assays for hazardous components of air pollution. Free Radic. Biol. Med. 117:202–17
    [Google Scholar]
  59. Fragou D, Pakkidi E, Aschner M, Samanidou V, Kovatsi L 2019. Smoking and DNA methylation: correlation of methylation with smoking behavior and association with diseases and fetus development following prenatal exposure. Food Chem. Toxicol. 129:312–27
    [Google Scholar]
  60. Frank JJ, Poulakos AG, Tornero-Velez R, Xue J 2019. Systematic review and meta-analyses of lead (Pb) concentrations in environmental media (soil, dust, water, food, and air) reported in the United States from 1996 to 2016. Sci. Total Environ. 694:133489
    [Google Scholar]
  61. GBD 2015 Tob. Collab 2017. Smoking prevalence and attributable disease burden in 195 countries and territories, 1990–2015: a systematic analysis from the Global Burden of Disease Study 2015. Lancet 389:1885–906
    [Google Scholar]
  62. Gibson L, Porter M. 2018. Drinking or smoking while breastfeeding and later cognition in children. Pediatrics 142:2e20174266
    [Google Scholar]
  63. Godfrey ME, Wojcik DP, Krone CA 2003. Apolipoprotein E genotyping as a potential biomarker for mercury neurotoxicity. J. Alzheimers Dis. 5:189–95
    [Google Scholar]
  64. Golding J, Gregory S, Iles-Caven Y, Nowicki S 2017. The mid-childhood and adolescent antecedents of women's external locus of control orientation. Wellcome Open Res 2:53
    [Google Scholar]
  65. Greer EL, Maures TJ, Ucar D, Hauswirth AG, Mancini E et al. 2011. Transgenerational epigenetic inheritance of longevity in Caenorhabditis elegans. Nature 479:365–71
    [Google Scholar]
  66. Gu H, Robison G, Hong L, Barrea R, Wei X et al. 2012. Increased β-amyloid deposition in Tg-SWDI transgenic mouse brain following in vivo lead exposure. Toxicol. Lett. 213:211–19
    [Google Scholar]
  67. Gumusoglu SB, Hing BWQ, Chilukuri ASS, Dewitt JJ, Scroggins SM, Stevens HE 2020. Chronic maternal interleukin-17 and autism-related cortical gene expression, neurobiology, and behavior. Neuropsychopharmacology 45:1008–17
    [Google Scholar]
  68. Guxens M, Lubczyńska MJ, Muetzel RL, Dalmau-Bueno A, Jaddoe VWV et al. 2018. Air pollution exposure during fetal life, brain morphology, and cognitive function in school-age children. Biol. Psychiatry 84:295–303
    [Google Scholar]
  69. Haghani A, Johnson R, Safi N, Zhang H, Thorwald M et al. 2020a. Toxicity of urban air pollution particulate matter in developing and adult mouse brain: comparison of total and filter-eluted nanoparticles. Environ. Int. 136:105510
    [Google Scholar]
  70. Haghani A, Johnson RG, Woodward NC, Feinberg JI, Lewis K et al. 2020b. Adult mouse hippocampal transcriptome changes associated with long-term behavioral and metabolic effects of gestational air pollution toxicity. Transl. Psychiatry 10:1218
    [Google Scholar]
  71. Hajat A, Hsia C, O'Neill MS 2015. Socioeconomic disparities and air pollution exposure: a global review. Curr. Environ. Health Rep. 2:440–50
    [Google Scholar]
  72. Hartman JD, Craig BM. 2018. Examining the association between maternal smoking during pregnancy and child behavior problems using quality-adjusted life years. Matern. Child Health J. 22:1780–88
    [Google Scholar]
  73. Hawkey A, Junaid S, Yao L, Spiera Z, White H et al. 2019. Gestational exposure to nicotine and/or benzo[a]pyrene causes long-lasting neurobehavioral consequences. Birth Defects Res 111:1248–58
    [Google Scholar]
  74. Hecht S. 2012. Research opportunities related to establishing standards for tobacco products under the Family Smoking Prevention and Tobacco Control Act. Nicotine Tob. Res. 14:18–28
    [Google Scholar]
  75. Heinonen K, Eriksson JG, Lahti J, Kajantie E, Pesonen AK et al. 2015. Late preterm birth and neurocognitive performance in late adulthood: a birth cohort study. Pediatrics 135:e818–25
    [Google Scholar]
  76. Homa DM, Neff LJ, King BA, Caraballo RS, Bunnell RE et al. 2015. Vital signs: disparities in nonsmokers’ exposure to secondhand smoke—United States, 1999–2012. Morb. Mortal. Wkly. Rep. 64:103–8
    [Google Scholar]
  77. Inst. Health Metrics Eval 2018. GBD compare data visualization Inst. Health Metr. Eval., Univ. Wash., Seattle WA, accessed Nov. 8. http://www.healthdata.org/data-visualization/gbd-compare
  78. Intern. Agency Res. Cancer 2004. IARC Monographs on the Evaluation of Carcinogenic Risks to Humans, Vol. 83 Tobacco Smoke and Involuntary Smoking Lyon, France: Intern. Agency Res. Cancer
  79. Jack CR Jr., Bennett DA, Blennow K, Carrillo MC, Dunn B et al. 2018. NIA-AA research framework: toward a biological definition of Alzheimer's disease. Alzheimer's Dement 14:535–62
    [Google Scholar]
  80. Jaddoe VW, de Ridder MA, van den Elzen AP, Hofman A, Uiterwaal CS, Witteman JC 2008a. Maternal smoking in pregnancy is associated with cholesterol development in the offspring: a 27-years follow-up study. Atherosclerosis 196:42–48
    [Google Scholar]
  81. Jaddoe VW, Troe EJ, Hofman A, Mackenbach JP, Moll HA et al. 2008b. Active and passive maternal smoking during pregnancy and the risks of low birthweight and preterm birth: the Generation R Study. Paediatr. Perinat. Epidemiol. 22:162–71
    [Google Scholar]
  82. Jiang C, Stewart LT, Kuo HC, McGilberry W, Wall SB et al. 2019. Cyclic O3 exposure synergizes with aging leading to memory impairment in male APOE ε3, but not APOE ε4, targeted replacement mice. Neurobiol. Aging 81:9–21
    [Google Scholar]
  83. Jo H, Eckel SP, Wang X, Chen JC, Cockburn M et al. 2019. Sex-specific associations of autism spectrum disorder with residential air pollution exposure in a large Southern California pregnancy cohort. Environ. Pollut. 254:Part A113010
    [Google Scholar]
  84. Johnson MD, Schilz J, Djordjevic MV, Rice JR, Shields PG 2009. Evaluation of in vitro assays for assessing the toxicity of cigarette smoke and smokeless tobacco. Cancer Epidemiol. Biomark. Prev. 18:3263–304
    [Google Scholar]
  85. Joubert BR, Håberg SE, Nilsen RM, Wang X, Vollset SE et al. 2012. 450K epigenome-wide scan identifies differential DNA methylation in newborns related to maternal smoking during pregnancy. Env. Health Perspect. 120:1425–31
    [Google Scholar]
  86. Kalkbrenner AE, Meier SM, Madley-Dowd P, Ladd-Acosta C, Fallin MD et al. 2020. Familial confounding of the association between maternal smoking in pregnancy and autism spectrum disorder in offspring. Autism Res 13:134–44
    [Google Scholar]
  87. Kim HW, Kam S, Lee DH 2014. Synergistic interaction between polycyclic aromatic hydrocarbons and environmental tobacco smoke on the risk of obesity in children and adolescents: The U.S. National Health and Nutrition Examination Survey 2003–2008. Environ. Res. 135:354–60
    [Google Scholar]
  88. Klocke C, Allen JL, Sobolewski M, Blum JL, Zelikoff JT, Cory-Slechta DA 2018a. Exposure to fine and ultrafine particulate matter during gestation alters postnatal oligodendrocyte maturation, proliferation capacity, and myelination. Neurotoxicology 65:196–206
    [Google Scholar]
  89. Klocke C, Allen JL, Sobolewski M, Mayer-Pröschel M, Blum JL et al. 2017. Neuropathological consequences of gestational exposure to concentrated ambient fine and ultrafine particles in the mouse. Toxicol. Sci. 156:492–508
    [Google Scholar]
  90. Klocke C, Sherina V, Graham UM, Gunderson J, Allen JL et al. 2018b. Enhanced cerebellar myelination with concomitant iron elevation and ultrastructural irregularities following prenatal exposure to ambient particulate matter in the mouse. Inhal. Toxicol. 30:381–96
    [Google Scholar]
  91. Kristjansson AL, Thomas S, Lilly CL, Thorisdottir IE, Allegrante JP, Sigfusdottir ID 2018. Maternal smoking during pregnancy and academic achievement of offspring over time: a registry data-based cohort study. Prev. Med. 113:74–79
    [Google Scholar]
  92. Kuhn HG, Dickinson-Anson H, Gage FH 1996. Neurogenesis in the dentate gyrus of the adult rat: age-related decrease of neuronal progenitor proliferation. J. Neurosci. 16:2027–33
    [Google Scholar]
  93. Kulick ER, Elkind MSV, Boehme AK, Joyce NR, Schupf N et al. 2020. Long-term exposure to ambient air pollution, APOE-ε4 status, and cognitive decline in a cohort of older adults in northern Manhattan. Environ. Int. 136:105440
    [Google Scholar]
  94. Küpers LK, Monnereau C, Sharp GC, Yousefi P, Salas LA et al. 2019. Meta-analysis of epigenome-wide association studies in neonates reveals widespread differential DNA methylation associated with birthweight. Nat. Commun. 10:1893
    [Google Scholar]
  95. Ladd-Acosta C, Feinberg JI, Brown SC, Lurmann FW, Croen LA et al. 2019. Epigenetic marks of prenatal air pollution exposure found in multiple tissues relevant for child health. Environ. Int. 126:363–76
    [Google Scholar]
  96. Landrigan PJ, Fuller R, Acosta NJR, Adeyi O, Arnold R et al. 2018. The Lancet Commission on pollution and health. Lancet 391:462–512
    [Google Scholar]
  97. Lange S, Probst C, Rehm J, Popova S 2018. National, regional, and global prevalence of smoking during pregnancy in the general population: a systematic review and meta-analysis. Lancet Glob. Health 6:e769–76
    [Google Scholar]
  98. Lavigne E, Ashley-Martin J, Dodds L, Arbuckle TE, Hystad P et al. 2016. Air pollution exposure during pregnancy and fetal markers of metabolic function: the MIREC study. Am. J. Epidemiol. 183:842–51
    [Google Scholar]
  99. Leggett RW. 1993. An age-specific kinetic model of lead metabolism in humans. Environ. Health Perspect. 101:598–616
    [Google Scholar]
  100. Leonardi-Bee J, Jere ML, Britton J 2011. Exposure to parental and sibling smoking and the risk of smoking uptake in childhood and adolescence: a systematic review and meta-analysis. Thorax 66:847–55
    [Google Scholar]
  101. Levy RJ. 2017. Carbon monoxide and anesthesia-induced neurotoxicity. Neurotoxicol. Teratol. 60:50–58
    [Google Scholar]
  102. Lotfipour S, Ferguson E, Leonard G, Miettunen J, Perron M et al. 2014. Maternal cigarette smoking during pregnancy predicts drug use via externalizing behavior in two community-based samples of adolescents. Addiction 109:1718–29
    [Google Scholar]
  103. Lubczyńska MJ, Muetzel RL, El Marroun H, Basagaña X, Strak M et al. 2020. Exposure to air pollution during pregnancy and childhood, and white matter microstructure in preadolescents. Environ. Health Perspect. 128:27005
    [Google Scholar]
  104. Malley CS, Kuylenstierna JC, Vallack HW, Henze DK, Blencowe H, Ashmore MR 2017. Preterm birth associated with maternal fine particulate matter exposure: a global, regional and national assessment. Environ. Int. 101:173–82
    [Google Scholar]
  105. Mao G, Nachman RM, Sun Q, Zhang X, Koehler K et al. 2017. Individual and joint effects of early-life ambient PM2.5 exposure and maternal prepregnancy obesity on childhood overweight or obesity. Environ. Health Perspect. 125:067005
    [Google Scholar]
  106. Marshall AT, Betts S, Kan EC, McConnell R, Lanphear BP, Sowell ER 2020. Association of lead-exposure risk and family income with childhood brain outcomes. Nat. Med. 26:91–97
    [Google Scholar]
  107. Martin EM, Fry RC. 2018. Environmental influences on the epigenome: exposure-associated DNA methylation in human populations. Annu. Rev. Public Health 39:309–33
    [Google Scholar]
  108. McCallister MM, Maguire M, Ramesh A, Aimin Q, Liu S et al. 2008. Prenatal exposure to benzo(a)pyrene impairs later-life cortical neuronal function. Neurotoxicology 29:846–54
    [Google Scholar]
  109. McConnell R, Shen E, Gilliland FD, Jerrett M, Wolch J et al. 2015. A longitudinal cohort study of body mass index and childhood exposure to secondhand tobacco smoke and air pollution: the Southern California Children's Health Study. Environ. Health Perspect 123:360–66 Erratum. 2015. Environ. Health Perspect. 123:A81
    [Google Scholar]
  110. McGuinn LA, Windham GC, Kalkbrenner AE, Bradley C, Di Q et al. 2020. Early life exposure to air pollution and autism spectrum disorder: findings from a multisite case-control study. Epidemiology 31:103–14
    [Google Scholar]
  111. Meleleo D, Notarachille G, Mangini V, Arnesano F 2019. Concentration-dependent effects of mercury and lead on Aβ42: possible implications for Alzheimer's disease. Eur. Biophys. J. 48:173–87
    [Google Scholar]
  112. Misra C, Kim S, Shen S, Sioutas C 2002. A high flow rate, very low pressure drop impactor for inertial separation of ultrafine from accumulation mode particles. J. Aerosol. Sci. 33:735–52
    [Google Scholar]
  113. Moore BF, Shapiro AL, Wilkening G, Magzamen S, Starling AP et al. 2020. Prenatal exposure to tobacco and offspring neurocognitive development in the Healthy Start study. J. Pediatr. 218:28–34
    [Google Scholar]
  114. Moreno-Gonzalez I, Estrada LD, Sanchez-Mejias E, Soto C 2013. Smoking exacerbates amyloid pathology in a mouse model of Alzheimer's disease. Nat. Commun. 4:1495
    [Google Scholar]
  115. Morgan TE, Davis DA, Iwata N, Tanner JA, Snyder D et al. 2011. Glutamatergic neurons in rodent models respond to nanoscale particulate urban air pollutants in vivo and in vitro. Environ. Health Perspect. 119:1003–9
    [Google Scholar]
  116. Mørkve Knudsen GT, Rezwan FI, Johannessen A, Skulstad SM, Bertelsen RJ et al. 2019. Epigenome-wide association of father's smoking with offspring DNA methylation: a hypothesis-generating study. Environ. Epigenet. 5:dvz023
    [Google Scholar]
  117. Mumaw CL, Levesque S, McGraw C, Robertson S, Lucas S et al. 2016. Microglial priming through the lung-brain axis: the role of air pollution-induced circulating factors. FASEB J 30:1880–91
    [Google Scholar]
  118. Musiek ES, Holtzman DM. 2015. Three dimensions of the amyloid hypothesis: time, space and ‘wingmen’. Nat. Neurosci. 18:800–6
    [Google Scholar]
  119. Nägga K, Gustavsson AM, Stomrud E, Lindqvist D, van Westen D et al. 2018. Increased midlife triglycerides predict brain β-amyloid and tau pathology 20 years later. Neurology 90:e73–81
    [Google Scholar]
  120. Natl. Acad. Sci. Eng. Med 2017. Using 21st Century Science to Improve Risk-Related Evaluations Washington, DC: Natl. Acad. Press
  121. Neal RE, Chen J, Jagadapillai R, Jang H, Abomoelak B et al. 2014. Developmental cigarette smoke exposure: hippocampus proteome and metabolome profiles in low birth weight pups. Toxicology 317:40–49
    [Google Scholar]
  122. Neal RE, Jagadapillai R, Chen J, Webb C, Stocke K et al. 2016. Developmental cigarette smoke exposure II: hippocampus proteome and metabolome profiles in adult offspring. Reprod. Toxicol. 65:436–47
    [Google Scholar]
  123. Obel C, Linnet KM, Henriksen TB, Rodriguez A, Järvelin MR et al. 2009. Smoking during pregnancy and hyperactivity-inattention in the offspring—comparing results from three Nordic cohorts. Int. J. Epidemiol. 38:698–705
    [Google Scholar]
  124. Ogliari KS, Lichtenfels AJ, de Marchi MR, Ferreira AT, Dolhnikoff M, Saldiva PH 2013. Intrauterine exposure to diesel exhaust diminishes adult ovarian reserve. Fertil. Steril. 99:1681–88
    [Google Scholar]
  125. Olmedo P, Goessler W, Tanda S, Grau-Perez M, Jarmul S et al. 2018. Metal concentrations in e-cigarette liquid and aerosol samples: the contribution of metallic coils. Environ. Health Perspect. 126:027010
    [Google Scholar]
  126. Oudin A, Frondelius K, Haglund N, Källén K, Forsberg B et al. 2019. Prenatal exposure to air pollution as a potential risk factor for autism and ADHD. Environ. Int. 133:Part A105149
    [Google Scholar]
  127. Palmer RH, Bidwell LC, Heath AC, Brick LA, Madden PA, Knopik VS 2016. Effects of maternal smoking during pregnancy on offspring externalizing problems: contextual effects in a sample of female twins. Behav. Genet. 46:403–15
    [Google Scholar]
  128. Peng L, Bonaguidi MA. 2018. Function and dysfunction of adult hippocampal neurogenesis in regeneration and disease. Am. J. Pathol. 188:23–28
    [Google Scholar]
  129. Perkins M, Wright RO, Amarasiriwardena CJ, Jayawardene I, Rifas-Shiman SL, Oken E 2014. Very low maternal lead level in pregnancy and birth outcomes in an eastern Massachusetts population. Ann. Epidemiol. 24:915–19
    [Google Scholar]
  130. Peterson BS, Rauh VA, Bansal R, Hao X, Toth Z et al. 2015. Effects of prenatal exposure to air pollutants (polycyclic aromatic hydrocarbons) on the development of brain white matter, cognition, and behavior in later childhood. JAMA Psychiatry 72:6531–40 Erratum. 2015. JAMA Psychiatry 72(6):625
    [Google Scholar]
  131. Polli FS, Ipsen TH, Caballero-Puntiverio M, Østerbøg TB, Aznar S et al. 2020. Cellular and molecular changes in hippocampal glutamate signaling and alterations in learning, attention, and impulsivity following prenatal nicotine exposure. Mol. Neurobiol. 57:2002–20
    [Google Scholar]
  132. Prada D, Colicino E, Power MC, Weisskopf MG, Zhong J et al. 2016. APOE ε4 allele modifies the association of lead exposure with age-related cognitive decline in older individuals. Environ. Res. 151:101–5
    [Google Scholar]
  133. Pryor WA. 1997. Cigarette smoke radicals and the role of free radicals in chemical carcinogenicity. Environ. Health Perspect. 105:875–82
    [Google Scholar]
  134. Reed MD, Yim YS, Wimmer RD, Kim H, Ryu C et al. 2020. IL-17a promotes sociability in mouse models of neurodevelopmental disorders. Nature 577:249–53
    [Google Scholar]
  135. Reis CF, de Souza ID, Morais DAA, Oliveira RAC, Imparato DO et al. 2019. Systems biology-based analysis indicates global transcriptional impairment in lead-treated human neural progenitor cells. Front. Genet. 10:791
    [Google Scholar]
  136. Reitz C. 2013. Dyslipidemia and the risk of Alzheimer's disease. Curr. Atheroscler. Rep. 15:307
    [Google Scholar]
  137. Reuben A, Caspi A, Belsky DW, Broadbent J, Harrington H et al. 2017. Association of childhood blood lead levels with cognitive function and socioeconomic status at age 38 years and with IQ change and socioeconomic mobility between childhood and adulthood. JAMA 317:1244–51
    [Google Scholar]
  138. Reuben A, Schaefer JD, Moffitt TE, Broadbent J, Harrington H et al. 2019. Association of childhood lead exposure with adult personality traits and lifelong mental health. JAMA Psychiatry 76:418–25
    [Google Scholar]
  139. Rice DC. 1992. Effect of lead during different developmental periods in the monkey on concurrent discrimination performance. Neurotoxicology 13:583–92
    [Google Scholar]
  140. Rich DQ, Liu K, Zhang J, Thurston SW, Stevens TP et al. 2015. Differences in birth weight associated with the 2008 Beijing Olympics air pollution reduction: results from a natural experiment. Environ. Health Perspect. 123:880–87
    [Google Scholar]
  141. Rosa MJ, Hair GM, Just AC, Kloog I, Svensson K et al. 2020. Identifying critical windows of prenatal particulate matter (PM2.5) exposure and early childhood blood pressure. Environ. Res. 182:109073
    [Google Scholar]
  142. Saenen ND, Martens DS, Neven KY, Alfano R, Bové H et al. 2019. Air pollution-induced placental alterations: an interplay of oxidative stress, epigenetics, and the aging phenotype?. Clin. Epigenetics 11:124
    [Google Scholar]
  143. Saenen ND, Plusquin M, Bijnens E, Janssen BG, Gyselaers W et al. 2015. In utero fine particle air pollution and placental expression of genes in the brain-derived neurotrophic factor signaling pathway: an ENVIRONAGE Birth Cohort Study. Environ. Health Perspect. 123:834–40
    [Google Scholar]
  144. Saenen ND, Vrijens K, Janssen BG, Madhloum N, Peusens M et al. 2016. Placental nitrosative stress and exposure to ambient air pollution during gestation: a population study. Am. J. Epidemiol. 184:442–49
    [Google Scholar]
  145. Saenen ND, Vrijens K, Janssen BG, Roels HA, Neven KY et al. 2017. Lower placental leptin promoter methyl-ation in association with fine particulate matter air pollution during pregnancy and placental nitrosative stress at birth in the ENVIRONAGE cohort. Environ. Health Perspect. 125:262–68
    [Google Scholar]
  146. Salminen LE, Wilcox RR, Zhu AH, Riedel BC, Ching CRK et al. 2019. Altered cortical brain structure and increased risk for disease seen decades after perinatal exposure to maternal smoking: a study of 9000 adults in the UK Biobank. Cereb. Cortex 29:5217–33
    [Google Scholar]
  147. Shirmohammadi F, Hasheminassab S, Wang D, Saffari A, Schauer JJ et al. 2015. Oxidative potential of coarse particulate matter PM10–2.5 and its relation to water solubility and sources of trace elements and metals in the Los Angeles Basin. Env. Sci. Process. Impacts 17:2110–21
    [Google Scholar]
  148. Shirmohammadi F, Hasheminassab S, Wang D, Schauer JJ, Shafer MM et al. 2016. The relative importance of tailpipe and non-tailpipe emissions on the oxidative potential of ambient particles in Los Angeles, CA. Faraday Discuss 189:361–80
    [Google Scholar]
  149. Signes-Pastor AJ, Doherty BT, Romano ME, Gleason KM, Gui J et al. 2019. Prenatal exposure to metal mixture and sex-specific birth outcomes in the New Hampshire Birth Cohort Study. Environ. Epidemiol. 3:e068
    [Google Scholar]
  150. Silverberg O, Park AL, Cohen E, Fell DB, Ray JG 2018. Premature cardiac disease and death in women whose infant was preterm and small for gestational age: a retrospective cohort study. JAMA Cardiol 3:247–51
    [Google Scholar]
  151. Simpson J, Smith AD, Fraser A, Sattar N, Lindsay RS et al. 2017. Programming of adiposity in childhood and adolescence: associations with birth weight and cord blood adipokines. J. Clin. Endocrinol. Metab. 102:499–506
    [Google Scholar]
  152. Singh G, Singh V, Sobolewski M, Cory-Slechta DA, Schneider JS 2018. Sex-dependent effects of developmental lead exposure on the brain. Front. Genet. 9:89
    [Google Scholar]
  153. Sobolewski M, Abston K, Conrad K, Marvin E, Harvey K et al. 2020. Lineage- and sex-dependent behavioral and biochemical transgenerational consequences of developmental exposure to lead, prenatal stress, and combined lead and prenatal stress in mice. Environ. Health Perspect. 128:27001
    [Google Scholar]
  154. Sobolewski M, Varma G, Adams B, Anderson DW, Schneider JS, Cory-Slechta DA 2018. Developmental lead exposure and prenatal stress result in sex-specific reprograming of adult stress physiology and epigenetic profiles in brain. Toxicol. Sci. 163:478–89
    [Google Scholar]
  155. Stelzmann RA, Schnitzlein HN, Murtagh FR 1995. An English translation of Alzheimer's 1907 paper, “Uber eine eigenartige Erkankung der Hirnrinde.”. Clin. Anat. 8:429–31
    [Google Scholar]
  156. Stroud LR, McCallum M, Salisbury AL 2018. Impact of maternal prenatal smoking on fetal to infant neurobehavioral development. Dev. Psychopathol. 30:1087–105
    [Google Scholar]
  157. Stroud LR, Papandonatos GD, Rodriguez D, McCallum M, Salisbury AL et al. 2014. Maternal smoking during pregnancy and infant stress response: test of a prenatal programming hypothesis. Psychoneuroendocrinology 48:29–40
    [Google Scholar]
  158. Sweeney MD, Montagne A, Sagare AP, Nation DA, Schneider LS et al. 2019. Vascular dysfunction—the disregarded partner of Alzheimer's disease. Alzheimer's Dement 15:158–67
    [Google Scholar]
  159. Taylor CM, Golding J, Emond AM 2014. Adverse effects of maternal lead levels on birth outcomes in the ALSPAC study: a prospective birth cohort study. BJOG 122:322–28
    [Google Scholar]
  160. Tena A, Peru E, Martinetti LE, Cano JC, Loyola Baltazar CD et al. 2019. Long-term consequences of early postnatal lead exposure on hippocampal synaptic activity in adult mice. Brain Behav 9:e01307
    [Google Scholar]
  161. Thakur GA, Sengupta SM, Grizenko N, Schmitz N, Pagé V, Joober R 2013. Maternal smoking during pregnancy and ADHD: a comprehensive clinical and neurocognitive characterization. Nicotine Tob. Res. 15:149–57
    [Google Scholar]
  162. Torres LH, Garcia RC, Blois AM, Dati LM, Durão AC et al. 2015. Exposure of neonatal mice to tobacco smoke disturbs synaptic proteins and spatial learning and memory from late infancy to early adulthood. PLOS ONE 10:e0136399
    [Google Scholar]
  163. Trumble B, Finch CE. 2019. The exposome in human evolution: from dust to diesel. Q. Rev. Biol. 94:333–94
    [Google Scholar]
  164. Turner MC, Cohen A, Burnett RT, Jerrett M, Diver WR et al. 2017. Interactions between cigarette smoking and ambient PM2.5 for cardiovascular mortality. Environ. Res. 154:304–10
    [Google Scholar]
  165. Turner MC, Cohen A, Jerrett M, Gapstur SM, Diver WR et al. 2014. Interactions between cigarette smoking and fine particulate matter in the Risk of Lung Cancer Mortality in Cancer Prevention Study II. Am. J. Epidemiol. 180:1145–49
    [Google Scholar]
  166. US Dep. Health Hum. Serv 2016. E-cigarette use among youth and young adults: a report of the Surgeon General US Dep. Health Hum. Serv., Cent. Dis. Control Prev. Atlanta, GA:
    [Google Scholar]
  167. Volk HE, Hertz-Picciotto I, Delwiche L, Lurmann F, McConnell R 2011. Residential proximity to freeways and autism in the CHARGE study. Environ. Health Perspect. 119:873–77
    [Google Scholar]
  168. Wang H, Li X, Guo J, Peng B, Cui H et al. 2016. Distribution of toxic chemicals in particles of various sizes from mainstream cigarette smoke. Inhal. Toxicol. 28:89–94
    [Google Scholar]
  169. Weintraub S, Dikmen SS, Heaton RK, Tulsky DS, Zelazo PD et al. 2013. Cognition assessment using the NIH Toolbox. Neurology 80:11 Suppl. 3S54–64
    [Google Scholar]
  170. Weisskopf MG, Wright RO, Schwartz J, Spiro A 3rd, Sparrow D et al. 2004. Cumulative lead exposure and prospective change in cognition among elderly men: the VA Normative Aging Study. Am. J. Epidemiol. 160:1184–93
    [Google Scholar]
  171. Weston HI, Weston DD, Allen JL, Cory-Slechta DA 2014. Sex-dependent impacts of low-level lead exposure and prenatal stress on impulsive choice behavior and associated biochemical and neurochemical manifestations. Neurotoxicology 44:169–83
    [Google Scholar]
  172. WHO (World Health Organ.) 2018. Household air pollution and health News Release May 8. https://www.who.int/news-room/fact-sheets/detail/household-air-pollution-and-health
  173. Wild CP 2012. The exposome: from concept to utility. Int. J. Epidemiol. 41:24–32
    [Google Scholar]
  174. Woodward NC, Crow AL, Zhang Y, Epstein S, Hartiala J et al. 2019. Exposure to nanoscale particulate matter from gestation to adulthood impairs metabolic homeostasis in mice. Sci. Rep. 9:1816
    [Google Scholar]
  175. Woodward NC, Haghani A, Johnson RG, Hsu TM, Saffari A et al. 2018. Prenatal and early life exposure to air pollution induced hippocampal vascular leakage and impaired neurogenesis in association with behavioral deficits. Transl. Psychiatry 8:261
    [Google Scholar]
  176. Woodward NC, Levine MC, Haghani A, Shirmohammadi F, Saffari A et al. 2017a. Toll-like receptor 4 in glial inflammatory responses to air pollution in vitro and in vivo. J. Neuroinflammation 14:84
    [Google Scholar]
  177. Woodward NC, Pakbin P, Saffari A, Shirmohammadi F, Haghani A et al. 2017b. Traffic-related air pollution impact on mouse brain accelerates myelin and neuritic aging changes with specificity for CA1 neurons. Neurobiol. Aging 53:48–58
    [Google Scholar]
  178. Wright JP, Dietrich KN, Ris MD, Hornung RW, Wessel SD et al. 2008. Association of prenatal and childhood blood lead concentrations with criminal arrests in early adulthood. PLOS Med 5:e101
    [Google Scholar]
  179. Wu J, Basha MR, Brock B, Cox DP, Cardozo-Pelaez F et al. 2008. Alzheimer's disease (AD)-like pathology in aged monkeys after infantile exposure to environmental metal lead (Pb): evidence for a developmental origin and environmental link for AD. J. Neurosci. 28:3–9
    [Google Scholar]
  180. Xu Y, Wang W, Chen M, Zhou J, Huang X et al. 2019. Developmental programming of obesity by maternal exposure to concentrated ambient PM2.5 is maternally transmitted into the third generation in a mouse model. Part. Fibre Toxicol. 16:27
    [Google Scholar]
  181. Yan Y, Bazzano LA, Juonala M, Raitakari OT, Viikari JSA et al. 2019. Long-term burden of increased body mass index from childhood on adult dyslipidemia: the i3C Consortium study. J. Clin. Med. 8:101725
    [Google Scholar]
  182. Yeter D, Banks EC, Aschner M 2020. Disparity in risk factor severity for early childhood blood lead among predominantly African-American black children: the 1999 to 2010 US NHANES. Int. J. Environ. Res. Public Health 17:51552
    [Google Scholar]
  183. Yokota S, Oshio S, Moriya N, Takeda K 2016. Social isolation-induced territorial aggression in male offspring is enhanced by exposure to diesel exhaust during pregnancy. PLOS ONE 11:e0149737
    [Google Scholar]
  184. Zhang M, Mueller NT, Wang H, Hong X, Appel LJ, Wang X 2018. Maternal exposure to ambient particulate matter ≤2.5 μm during pregnancy and the risk for high blood pressure in childhood. Hypertension 72:194–201
    [Google Scholar]
/content/journals/10.1146/annurev-devpsych-042320-044338
Loading
/content/journals/10.1146/annurev-devpsych-042320-044338
Loading

Data & Media loading...

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error