Skip to main content

Advertisement

Log in

Early Environmental Exposures and Contaminants: a Design Framework for Biospecimen Collection and Analysis for a Prospective National Birth Cohort

  • ORIGINAL ARTICLE
  • Published:
Adversity and Resilience Science Aims and scope Submit manuscript

Abstract

Identifying factors associated with disruptions in early neurodevelopment is imperative for promoting the health and wellbeing of children. We describe a design framework for biospecimen collection for the forthcoming HEALthy Brain and Child Development (HBCD) study, which seeks to establish a large cohort of pregnant women throughout the USA and follow their children into middle childhood. Biospecimens and biological outcomes of interest, together with validated questionnaires and other measures, may help to disentangle the effect of prenatal exposures from social and environment factors that extend across the prenatal and postnatal periods, including early life adversity (ELA). Biospecimen selection is discussed across four domains of interest: (a) substance use exposure, (b) other environmental exposures, (c) genomics and epigenomics, and (d) other biological markers of neurodevelopment and putative moderators and mediators of developmental effects. HBCD biospecimen working group recommendations were based on nine guiding principles including utility as a biomarker to assess neurodevelopment; feasibility of collection during critical periods of exposure with a broad detection window; logistics and cost of specimen collection, storage, and transportation; cultural acceptability; minimal invasiveness; flexibility; high sensitivity and specificity; anticipated expertise and infrastructure at participating sites; and availability of informative alternative non-biological measures. The proposed essential and recommended biospecimens will need to be integrated with the recommendations from other HBCD working groups to ascertain overall burden for the research participants and families, feasibility of multi-modal collection at each study visit, cost, and implications for recruitment and retention, as well as any potential legal repercussions.

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.

Institutional subscriptions

Similar content being viewed by others

References

  • Abraham, M., Alramadhan, S., Iniguez, C., Duijts, L., Jaddoe, V. W., Den Dekker, H. T., et al. (2017). A systematic review of maternal smoking during pregnancy and fetal measurements with meta-analysis. PLoS One, 12(2), e0170946.

    PubMed  PubMed Central  Google Scholar 

  • Adamo, A. M., & Oteiza, P. I. (2010). Zinc deficiency and neurodevelopment: The case of neurons. Biofactors, 36(2), 117–124.

    PubMed  PubMed Central  Google Scholar 

  • Allen, L. H., Miller, J. W., de Groot, L., Rosenberg, I. H., Smith, A. D., Refsum, H., et al. (2018). Biomarkers of nutrition for development (BOND): Vitamin B-12 review. Journal of Nutrition, 148(suppl_4), 1995s–2027s.

    Google Scholar 

  • Anderson, O. S., Sant, K. E., & Dolinoy, D. C. (2012). Nutrition and epigenetics: An interplay of dietary methyl donors, one-carbon metabolism and DNA methylation. Journal of Nutritional Biochemistry, 23(8), 853–859.

    Google Scholar 

  • Apter-Levi, Y., Pratt, M., Vakart, A., Feldman, M., Zagoory-Sharon, O., & Feldman, R. (2016). Maternal depression across the first years of life compromises child psychosocial adjustment; relations to child HPA-axis functioning. Psychoneuroendocrinology, 64, 47–56.

    PubMed  Google Scholar 

  • Attramadal, A., & Jonsen, J. (1976). The content of lead, cadmium, zinc and copper in deciduous and permanent human teeth. Acta Odontology Scandanavia, 34(3), 127–131.

    Google Scholar 

  • Bailey, L. B., Stover, P. J., McNulty, H., Fenech, M. F., Gregory 3rd, J. F., Mills, J. L., et al. (2015). Biomarkers of nutrition for development-folate review. Journal of Nutrition, 145(7), 1636s–1680s.

    Google Scholar 

  • Bakhireva, L. N., Holbrook, B. D., Shrestha, S., Leyva, Y., Ashley, M., Cano, S., Lowe, J., Stephen, J. M., & Leeman, L. (2019). Association between prenatal opioid exposure, neonatal opioid withdrawal syndrome, and neurodevelopmental and behavioral outcomes at 5-8 months of age. Early Human Development, 128, 69–76.

    PubMed  Google Scholar 

  • Bakhireva, L. N., Nebeker, C., Ossorio, P., Angal, J., Thomason, M. E., & Croff, J. M. (2020). Inclusion of native Americans and Alaskan natives in large national studies: Ethical considerations and implications for biospecimen collection in the HEALthy brain and child development study. Adversity and Resilience Science.

  • Bakhireva, L. N., Shrestha, S., Garrison, L., Leeman, L., Rayburn, W. F., & Stephen, J. M. (2018). Prevalence of alcohol use in pregnant women with substance use disorder. Drug and Alcohol Dependence, 187, 305–310.

    PubMed  PubMed Central  Google Scholar 

  • Barker, D. J. (2004). The developmental origins of chronic adult disease. Acta Paediatrica. Supplement, 93(446), 26–33.

    Google Scholar 

  • Baumeister, D., Akhtar, R., Ciufolini, S., Pariante, C. M., & Mondelli, V. (2016). Childhood trauma and adulthood inflammation: A meta-analysis of peripheral C-reactive protein, interleukin-6 and tumour necrosis factor-α. Molecular Psychiatry, 21(5), 642–649.

    PubMed  Google Scholar 

  • Beauchamp, K. G., Lowe, J., Schrader, R. M., Shrestha, S., Aragón, C., Moss, N., Stephen, J. M., & Bakhireva, L. N. (2020). Self-regulation and emotional reactivity in infants with prenatal exposure to opioids and alcohol. Early Human Development, 148, 105119.

    PubMed  Google Scholar 

  • Bergamasco, L., Macchi, E., Facello, C., Badino, P., Odore, R., Pagliasso, S., Bellino, C., Osella, M. C., & Re, G. (2005). Effects of brief maternal separation in kids on neurohormonal and electroencephalographic parameters. Applied Animal Behaviour Science, 93(1–2), 39–52.

    Google Scholar 

  • Bergman, K., Glover, V., Sarkar, P., Abbott, D. H., & O'Connor, T. G. (2010). In utero cortisol and testosterone exposure and fear reactivity in infancy. Hormones and Behavior, 57(3), 306–312.

    PubMed  PubMed Central  Google Scholar 

  • Bjork, J. M., Straub, L. K., Provost, R. G., & Neale, M. C. (2017). The ABCD study of neurodevelopment: Identifying neurocircuit targets for prevention and treatment of adolescent substance abuse. Current Treatment Options in Psychiatry, 4(2), 196–209.

    PubMed  PubMed Central  Google Scholar 

  • Bogdan, R., Baranger, D. A. A., & Agrawal, A. (2018). Polygenic risk scores in clinical psychology: Bridging genomic risk to individual differences. Annual Review of Clinical Psychology, 14, 119–157.

    PubMed  Google Scholar 

  • Buck Louis, G. M., Smarr, M. M., & Patel, C. J. (2017). The exposome research paradigm: An opportunity to understand the environmental basis for human health and disease. Current Environmental Health Reports, 4(1), 89–98.

    PubMed  PubMed Central  Google Scholar 

  • Buckley JP, Barrett ES, Beamer PI, Bennett DH, Bloom MS, Fennell TR, Fry RC, Funk WE, Hamra GB, Hecht SS, Kannan K, Iyer R, Karagas MR, Lyall K, Parsons PJ, Pellizzari ED, Signes-Pastor AJ, Starling AP, Wang A, Watkins DJ, Zhang M, Woodruff TJ; program collaborators for ECHO. ( 2020)Opportunities for evaluating chemical exposures and child health in the United States: the Environmental influences on Child Health Outcomes (ECHO) Program. J Expo Sci Environ Epidemiol, 30(3):397–419. https://doi.org/10.1038/s41370-020-0211-9.

  • Buniello, A., MacArthur, J. A. L., Cerezo, M., Harris, L. W., Hayhurst, J., Malangone, C., et al. (2019). The NHGRIEBI GWAS Catalog of published genome-wide association studies, targeted arrays and summary statistics 2019. Nucleic Acids Res, 47(D1), D1005–D1012. https://doi.org/10.1093/nar/gky1120.

    Article  PubMed  Google Scholar 

  • Byun, K., Won, Y. L., Hwang, Y. I., Koh, D. H., Im, H., & Kim, E. A. (2013). Assessment of arsenic exposure by measurement of urinary speciated inorganic arsenic metabolites in workers in a semiconductor manufacturing plant. Annals of Occupational and Environmental Medicine, 25(1), 21.

    PubMed  PubMed Central  Google Scholar 

  • Camann, D. E., Schultz, S. T., Yau, A. Y., Heilbrun, L. P., Zuniga, M. M., Palmer, R. F., & Miller, C. S. (2013). Acetaminophen, pesticide, and diethylhexyl phthalate metabolites, anandamide, and fatty acids in deciduous molars: Potential biomarkers of perinatal exposure. Journal of Exposure Science & Environmental Epidemiology, 23(2), 190–196.

    Google Scholar 

  • Challis, J. R., Sloboda, D., Matthews, S. G., Holloway, A., Alfaidy, N., Patel, F. A., et al. (2001). The fetal placental hypothalamic-pituitary-adrenal (HPA) axis, parturition and post natal health. Molecular and Cellular Endocrinology, 185(1–2), 135–144.

    PubMed  Google Scholar 

  • Chapman, K., Holmes, M., & Seckl, J. (2013). 11β-hydroxysteroid dehydrogenases: Intracellular gate-keepers of tissue glucocorticoid action. Physiological Reviews, 93(3), 1139–1206.

    PubMed  PubMed Central  Google Scholar 

  • Chiodo, L. M., Janisse, J., Delaney-Black, V., Sokol, R. J., & Hannigan, J. H. (2009). A metric of maternal prenatal risk drinking predicts neurobehavioral outcomes in preschool children. Alcoholism, Clinical and Experimental Research, 33(4), 634–644.

    PubMed  Google Scholar 

  • Chuang, H.-Y., Schwartz, J., Gonzales-Cossio, T., Lugo, M. C., Palazuelos, E., Aro, A., Hu, H., & Hernandez-Avila, M. (2001). Interrelations of lead levels in bone, venous blood, and umbilical cord blood with exogenous lead exposure through maternal plasma lead in peripartum women. Environmental Health Perspectives, 109(5), 527–532.

    PubMed  PubMed Central  Google Scholar 

  • Colby, J. M. (2017). Comparison of umbilical cord tissue and meconium for the confirmation of in utero drug exposure. Clinical Biochemistry, 50(13–14), 784–790.

    PubMed  Google Scholar 

  • Comeau, W. L., Lee, K., Anderson, K., & Weinberg, J. (2015). Prenatal alcohol exposure and adolescent stress increase sensitivity to stress and gonadal hormone influences on cognition in adult female rats. Physiology and Behavior, 148, 157–165.

    PubMed  Google Scholar 

  • Cong, X., Xu, W., Janton, S., Henderson, W. A., Matson, A., McGrath, J. M., Maas, K., & Graf, J. (2016). Gut microbiome developmental patterns in early life of preterm infants: Impacts of feeding and gender. PLoS One, 11(4), e0152751.

    PubMed  PubMed Central  Google Scholar 

  • Corsi, D. J., Donelle, J., Sucha, E., Hawken, S., Hsu, H., El-Chaâr, D., et al. (2020). Maternal cannabis use in pregnancy and child neurodevelopmental outcomes. Nature Medicine, 26, 1536–1540.

    PubMed  Google Scholar 

  • Crawford, M., Doyle, W., Leaf, A., Leighfield, M., Ghebremeskel, K., & Phylactos, A. (1993). Nutrition and neurodevelopmental disorders. Nutrition and Health, 9(2), 81–97.

    PubMed  Google Scholar 

  • Cross-Disorder Group of the Psychiatric Genomics Consortium. (2019). Genomic relationships, novel loci, and pleiotropic mechanisms across eight psychiatric disorders. Cell, 179(7), 1469–1482.e1411.

    PubMed Central  Google Scholar 

  • Dahmen, B., Puetz, V. B., Scharke, W., von Polier, G. G., Herpertz-Dahlmann, B., & Konrad, K. (2018). Effects of early-life adversity on hippocampal structures and associated HPA axis functions. Developmental Neuroscience, 40(1), 13–22.

    PubMed  Google Scholar 

  • Davis, E. P., Glynn, L. M., Waffarn, F., & Sandman, C. A. (2011). Prenatal maternal stress programs infant stress regulation. Journal of Child Psychology and Psychiatry, 52(2), 119–129.

    PubMed  Google Scholar 

  • de Castro, A., Jones, H. E., Johnson, R. E., Gray, T. R., Shakleya, D. M., & Huestis, M. A. (2011). Methadone, cocaine, opiates, and metabolite disposition in umbilical cord and correlations to maternal methadone dose and neonatal outcomes. Therapeutic Drug Monitoring, 33(4), 443–452.

    PubMed  PubMed Central  Google Scholar 

  • DiNieri, J. A., Wang, X., Szutorisz, H., Spano, S. M., Kaur, J., Casaccia, P., Dow-Edwards, D., & Hurd, Y. L. (2011). Maternal cannabis use alters ventral striatal dopamine D2 gene regulation in the offspring. Biological Psychiatry, 70(8), 763–769.

    PubMed  PubMed Central  Google Scholar 

  • Dolan, K., Rouen, D., & Kimber, J. (2004). An overview of the use of urine, hair, sweat and saliva to detect drug use. Drug and Alcohol Review, 23(2), 213–217.

    PubMed  Google Scholar 

  • Dórea, J. G. (2019). Environmental exposure to low-level lead (Pb) co-occurring with other neurotoxicants in early life and neurodevelopment of children. Environmental Research, 177, 108641.

    PubMed  Google Scholar 

  • Eichner, E. R., & Hillman, R. S. (1973). Effect of alcohol on serum folate level. The Journal of Clinical Investigation, 52(3), 584–591.

    PubMed  PubMed Central  Google Scholar 

  • Ejaredar, M., Nyanza, E. C., Ten Eycke, K., & Dewey, D. (2015). Phthalate exposure and childrens neurodevelopment: A systematic review. Environmental Research, 142, 51–60.

    PubMed  Google Scholar 

  • Escher, B. I., Hackermüller, J., Polte, T., Scholz, S., Aigner, A., Altenburger, R., Böhme, A., Bopp, S. K., Brack, W., Busch, W., Chadeau-Hyam, M., Covaci, A., Eisenträger, A., Galligan, J. J., Garcia-Reyero, N., Hartung, T., Hein, M., Herberth, G., Jahnke, A., Kleinjans, J., Klüver, N., Krauss, M., Lamoree, M., Lehmann, I., Luckenbach, T., Miller, G. W., Müller, A., Phillips, D. H., Reemtsma, T., Rolle-Kampczyk, U., Schüürmann, G., Schwikowski, B., Tan, Y. M., Trump, S., Walter-Rohde, S., & Wambaugh, J. F. (2017). From the exposome to mechanistic understanding of chemical-induced adverse effects. Environment International, 99, 97–106.

    PubMed  Google Scholar 

  • Farzan, S. F., Karagas, M. R., & Chen, Y. (2013). In utero and early life arsenic exposure in relation to long-term health and disease. Toxicology and Applied Pharmacology, 272(2), 384–390.

    PubMed  PubMed Central  Google Scholar 

  • Fattorusso, A., Di Genova, L., Dell’Isola, G. B., Mencaroni, E., & Esposito, S. (2019). Autism spectrum disorders and the gut microbiota. Nutrients, 11(3), 521.

    PubMed Central  Google Scholar 

  • Flynn, A., Martier, S., Sokol, R., Miller, S., Golden, N., & Del Villano, B. (1981). Zinc status of pregnant alcoholic women: A determinant of fetal outcome. The Lancet, 317(8220), 572–575.

    Google Scholar 

  • Forney, L. J., Gajer, P., Williams, C. J., Schneider, G. M., Koenig, S. S., McCulle, S. L., et al. (2010). Comparison of self-collected and physician-collected vaginal swabs for microbiome analysis. Journal of Clinical Microbiology, 48(5), 1741–1748.

    PubMed  PubMed Central  Google Scholar 

  • Fried, P. A., Watkinson, B., & Gray, R. (1998). Differential effects on cognitive functioning in 9- to 12-year olds prenatally exposed to cigarettes and marihuana. Neurotoxicology and Teratology, 20(3), 293–306.

    PubMed  Google Scholar 

  • Gabriel, H. E., Crott, J. W., Ghandour, H., Dallal, G. E., Choi, S.-W., Keyes, M. K., Jang, H., Liu, Z., Nadeau, M., Johnston, A., Mager, D., & Mason, J. B. (2006). Chronic cigarette smoking is associated with diminished folate status, altered folate form distribution, and increased genetic damage in the buccal mucosa of healthy adults. The American Journal of Clinical Nutrition, 83(4), 835–841.

    PubMed  Google Scholar 

  • Galicia-García, V., Rojas-Lopez, M., Rojas, R., Olaiz, G., & Rios, C. (1997). Cadmium levels in maternal, cord and newborn blood in Mexico City. Toxicology Letters, 91(1), 57–61.

    PubMed  Google Scholar 

  • Galvin, S. L., Ramage, M., Mazure, E., & Coulson, C. C. (2020). The association of cannabis use late in pregnancy with engagement and retention in perinatal substance use disorder care for opioid use disorder: A cohort comparison. J Subst Abuse Treat, 117, 108098. https://doi.org/10.1016/j.jsat.2020.108098.

    Article  PubMed  Google Scholar 

  • Garg, M., Garrison, L., Leeman, L., Hamidovic, A., Borrego, M., Rayburn, W. F., & Bakhireva, L. (2016). Validity of self-reported drug use information among pregnant women. Maternal and Child Health Journal, 20(1), 41–47.

    PubMed  PubMed Central  Google Scholar 

  • Gatny, H. H., Couper, M. P., & Axinn, W. G. (2013). New strategies for biosample collection in population-based social research. Social Science Research, 42(5), 1402–1409.

    PubMed  PubMed Central  Google Scholar 

  • Girchenko, P., Lahti-Pulkkinen, M., Heinonen, K., Reynolds, R. M., Laivuori, H., Lipsanen, J., Villa, P. M., Hämäläinen, E., Kajantie, E., Lahti, J., & Räikkönen, K. (2020). Persistently high levels of maternal antenatal inflammation are associated with and mediate the effect of prenatal environmental adversities on neurodevelopmental delay in the offspring. Biological Psychiatry, 87(10), 898–907.

    PubMed  Google Scholar 

  • Gray, T. R., Choo, R. E., Concheiro, M., Williams, E., Elko, A., Jansson, L. M., Jones, H. E., & Huestis, M. A. (2010). Prenatal methadone exposure, meconium biomarker concentrations and neonatal abstinence syndrome. Addiction, 105(12), 2151–2159.

    PubMed  PubMed Central  Google Scholar 

  • Groves, R. M., Cialdini, R. B., & Couper, M. P. (1992). Understanding the decision to participate in a survey. Public Opinion Quarterly, 56(4), 475–495.

    Google Scholar 

  • Hadizadeh, H., Salehi, M., Bozorgnia, A. R., & Ahmadkhaniha, H. R. (2020). Lower folate levels in methamphetamine-induced psychosis: A cross-sectional study. Drug and Alcohol Dependence, 207, 107682.

    PubMed  Google Scholar 

  • Halpern, S. D. (2011). Financial incentives for research participation: Empirical questions, available answers and the burden of further proof. American Journal Medical Science, 342(4), 290–293.

    Google Scholar 

  • Hartwell, M. L., Croff, J. M., Morris, A. S., Breslin, F. J., & Dunn, K. (2020). Association of Prenatal Opioid Exposure With Precentral Gyrus Volume in Children. JAMA Pediatr, 174(9), 893–896. https://doi.org/10.1001/jamapediatrics.2020.0937.

    Article  PubMed  Google Scholar 

  • Herbstman, J. B., Sjödin, A., Kurzon, M., Lederman, S. A., Jones, R. S., Rauh, V., Needham, L. L., Tang, D., Niedzwiecki, M., Wang, R. Y., & Perera, F. (2010). Prenatal exposure to PBDEs and neurodevelopment. Environmental Health Perspectives, 118(5), 712–719.

    PubMed  PubMed Central  Google Scholar 

  • Hoffman, E. A., Clark, D. B., Orendain, N., Hudziak, J., Squeglia, L. M., & Dowling, G. J. (2019). Stress exposures, neurodevelopment and health measures in the ABCD study. Neurobiology of Stress, 10, 100157.

    PubMed  PubMed Central  Google Scholar 

  • Huizink, A. C. (2014). Prenatal cannabis exposure and infant outcomes: Overview of studies. Progress in Neuro-Psychopharmacology and Biological Psychiatry, 52, 45–52.

    PubMed  Google Scholar 

  • Jaddoe, V. W., van Duijn, C. M., Franco, O. H., van der Heijden, A. J., van Iizendoorn, M. H., de Jongste, J. C., et al. (2012). The generation R study: Design and cohort update 2012. European Journal of Epidemiology, 27(9), 739–756.

    PubMed  Google Scholar 

  • Javors, M. A., Hatch, J. P., & Lamb, R. J. (2011). Sequential combination of self-report, breath carbon monoxide, and saliva cotinine to assess smoking status. Drug and Alcohol Dependence, 113(2–3), 242–244.

    PubMed  Google Scholar 

  • Johnson, S. B., Riis, J. L., & Noble, K. G. (2016). State of the art review: Poverty and the developing brain. Pediatrics, 137(4), e20153075.

    PubMed  PubMed Central  Google Scholar 

  • Johnston, J. E., Franklin, M., Roh, H., Austin, C., & Arora, M. (2019). Lead and arsenic in shed deciduous teeth of children living near a lead-acid battery smelter. Environmental Science & Technology, 53(10), 6000–6006.

    Google Scholar 

  • Jones, A. W., & Andersson, L. (2003). Comparison of ethanol concentrations in venous blood and end-expired breath during a controlled drinking study. Forensic Science International, 132(1), 18–25.

    PubMed  Google Scholar 

  • Jones, H. E., Fischer, G., Heil, S. H., Kaltenbach, K., Martin, P. R., Coyle, M. G., et al. (2012). Maternal opioid treatment: Human experimental research (MOTHER)--approach, issues and lessons learned. Addiction, 107(Suppl 1(0 1)), 28–35.

    PubMed  PubMed Central  Google Scholar 

  • Kim, Y., Ha, E. H., Park, H., Ha, M., Kim, Y., Hong, Y. C., Kim, E. J., & Kim, B. N. (2013). Prenatal lead and cadmium co-exposure and infant neurodevelopment at 6 months of age: The mothers and children’s environmental health (MOCEH) study. Neurotoxicology, 35, 15–22.

    PubMed  Google Scholar 

  • King, J. C., Brown, K. H., Gibson, R. S., Krebs, N. F., Lowe, N. M., Siekmann, J. H., & Raiten, D. J. (2015). Biomarkers of nutrition for development (BOND)—Zinc review. The Journal of Nutrition, 146(4), 858S–885S.

    PubMed  Google Scholar 

  • Kippler, M., Tofail, F., Hamadani, J. D., Gardner, R. M., Grantham-McGregor, S. M., Bottai, M., & Vahter, M. (2012). Early-life cadmium exposure and child development in 5-year-old girls and boys: A cohort study in rural Bangladesh. Environmental Health Perspectives, 120(10), 1462–1468.

    PubMed  PubMed Central  Google Scholar 

  • Kong, A., Thorleifsson, G., Frigge, M. L., Vilhjalmsson, B. J., Young, A. I., Thorgeirsson, T. E., Benonisdottir, S., Oddsson, A., Halldorsson, B. V., Masson, G., Gudbjartsson, D. F., Helgason, A., Bjornsdottir, G., Thorsteinsdottir, U., & Stefansson, K. (2018). The nature of nurture: Effects of parental genotypes. Science, 359(6374), 424–428.

    PubMed  Google Scholar 

  • Kuhnle, G. G. (2012). Nutritional biomarkers for objective dietary assessment. Journal of the Science of Food and Agriculture, 92(6), 1145–1149.

    PubMed  Google Scholar 

  • Lammert, C. R., Frost, E. L., Bolte, A. C., Paysour, M. J., Shaw, M. E., Bellinger, C. E., Weigel, T. K., Zunder, E. R., & Lukens, J. R. (2018). Cutting edge: Critical roles for microbiota-mediated regulation of the immune system in a prenatal immune activation model of autism. The Journal of Immunology, 201(3), 845–850.

    PubMed  Google Scholar 

  • Lander, L., Howsare, J., & Byrne, M. (2013). The impact of substance use disorders on families and children: From theory to practice. Social Work in Public Health, 28(3–4), 194–205.

    PubMed  PubMed Central  Google Scholar 

  • Lecuyer, M., Laquerrière, A., Bekri, S., Lesueur, C., Ramdani, Y., Jégou, S., Uguen, A., Marcorelles, P., Marret, S., & Gonzalez, B. J. (2017). PLGF, a placental marker of fetal brain defects after in utero alcohol exposure. Acta Neuropathologica Communications, 5(1), 44.

    PubMed  PubMed Central  Google Scholar 

  • Ligthart, S., Vaez, A., Võsa, U., Stathopoulou, M. G., de Vries, P. S., Prins, B. P., van der Most, P. J., Tanaka, T., Naderi, E., Rose, L. M., Wu, Y., Karlsson, R., Barbalic, M., Lin, H., Pool, R., Zhu, G., Macé, A., Sidore, C., Trompet, S., Mangino, M., Sabater-Lleal, M., Kemp, J. P., Abbasi, A., Kacprowski, T., Verweij, N., Smith, A. V., Huang, T., Marzi, C., Feitosa, M. F., Lohman, K. K., Kleber, M. E., Milaneschi, Y., Mueller, C., Huq, M., Vlachopoulou, E., Lyytikäinen, L. P., Oldmeadow, C., Deelen, J., Perola, M., Zhao, J. H., Feenstra, B., Amini, M., Lahti, J., Schraut, K. E., Fornage, M., Suktitipat, B., Chen, W. M., Li, X., Nutile, T., Malerba, G., Luan, J.’., Bak, T., Schork, N., del Greco M., F., Thiering, E., Mahajan, A., Marioni, R. E., Mihailov, E., Eriksson, J., Ozel, A. B., Zhang, W., Nethander, M., Cheng, Y. C., Aslibekyan, S., Ang, W., Gandin, I., Yengo, L., Portas, L., Kooperberg, C., Hofer, E., Rajan, K. B., Schurmann, C., den Hollander, W., Ahluwalia, T. S., Zhao, J., Draisma, H. H. M., Ford, I., Timpson, N., Teumer, A., Huang, H., Wahl, S., Liu, Y. M., Huang, J., Uh, H. W., Geller, F., Joshi, P. K., Yanek, L. R., Trabetti, E., Lehne, B., Vozzi, D., Verbanck, M., Biino, G., Saba, Y., Meulenbelt, I., O’Connell, J. R., Laakso, M., Giulianini, F., Magnusson, P. K. E., Ballantyne, C. M., Hottenga, J. J., Montgomery, G. W., Rivadineira, F., Rueedi, R., Steri, M., Herzig, K. H., Stott, D. J., Menni, C., Frånberg, M., St. Pourcain, B., Felix, S. B., Pers, T. H., Bakker, S. J. L., Kraft, P., Peters, A., Vaidya, D., Delgado, G., Smit, J. H., Großmann, V., Sinisalo, J., Seppälä, I., Williams, S. R., Holliday, E. G., Moed, M., Langenberg, C., Räikkönen, K., Ding, J., Campbell, H., Sale, M. M., Chen, Y. D. I., James, A. L., Ruggiero, D., Soranzo, N., Hartman, C. A., Smith, E. N., Berenson, G. S., Fuchsberger, C., Hernandez, D., Tiesler, C. M. T., Giedraitis, V., Liewald, D., Fischer, K., Mellström, D., Larsson, A., Wang, Y., Scott, W. R., Lorentzon, M., Beilby, J., Ryan, K. A., Pennell, C. E., Vuckovic, D., Balkau, B., Concas, M. P., Schmidt, R., Mendes de Leon, C. F., Bottinger, E. P., Kloppenburg, M., Paternoster, L., Boehnke, M., Musk, A. W., Willemsen, G., Evans, D. M., Madden, P. A. F., Kähönen, M., Kutalik, Z., Zoledziewska, M., Karhunen, V., Kritchevsky, S. B., Sattar, N., Lachance, G., Clarke, R., Harris, T. B., Raitakari, O. T., Attia, J. R., van Heemst, D., Kajantie, E., Sorice, R., Gambaro, G., Scott, R. A., Hicks, A. A., Ferrucci, L., Standl, M., Lindgren, C. M., Starr, J. M., Karlsson, M., Lind, L., Li, J. Z., Chambers, J. C., Mori, T. A., de Geus, E. J. C. N., Heath, A. C., Martin, N. G., Auvinen, J., Buckley, B. M., de Craen, A. J. M., Waldenberger, M., Strauch, K., Meitinger, T., Scott, R. J., McEvoy, M., Beekman, M., Bombieri, C., Ridker, P. M., Mohlke, K. L., Pedersen, N. L., Morrison, A. C., Boomsma, D. I., Whitfield, J. B., Strachan, D. P., Hofman, A., Vollenweider, P., Cucca, F., Jarvelin, M. R., Jukema, J. W., Spector, T. D., Hamsten, A., Zeller, T., Uitterlinden, A. G., Nauck, M., Gudnason, V., Qi, L., Grallert, H., Borecki, I. B., Rotter, J. I., März, W., Wild, P. S., Lokki, M. L., Boyle, M., Salomaa, V., Melbye, M., Eriksson, J. G., Wilson, J. F., Penninx, B. W. J. H., Becker, D. M., Worrall, B. B., Gibson, G., Krauss, R. M., Ciullo, M., Zaza, G., Wareham, N. J., Oldehinkel, A. J., Palmer, L. J., Murray, S. S., Pramstaller, P. P., Bandinelli, S., Heinrich, J., Ingelsson, E., Deary, I. J., Mägi, R., Vandenput, L., van der Harst, P., Desch, K. C., Kooner, J. S., Ohlsson, C., Hayward, C., Lehtimäki, T., Shuldiner, A. R., Arnett, D. K., Beilin, L. J., Robino, A., Froguel, P., Pirastu, M., Jess, T., Koenig, W., Loos, R. J. F., Evans, D. A., Schmidt, H., Smith, G. D., Slagboom, P. E., Eiriksdottir, G., Morris, A. P., Psaty, B. M., Tracy, R. P., Nolte, I. M., Boerwinkle, E., Visvikis-Siest, S., Reiner, A. P., Gross, M., Bis, J. C., Franke, L., Franco, O. H., Benjamin, E. J., Chasman, D. I., Dupuis, J., Snieder, H., Dehghan, A., Alizadeh, B. Z., Alizadeh, B. Z., Boezen, H. M., Franke, L., van der Harst, P., Navis, G., Rots, M., Snieder, H., Swertz, M., Wolffenbuttel, B. H. R., Wijmenga, C., Benjamin, E., Chasman, D. I., Dehghan, A., Ahluwalia, T. S., Meigs, J., Tracy, R., Alizadeh, B. Z., Ligthart, S., Bis, J., Eiriksdottir, G., Pankratz, N., Gross, M., Rainer, A., Snieder, H., Wilson, J. G., Psaty, B. M., Dupuis, J., Prins, B., Vaso, U., Stathopoulou, M., Franke, L., Lehtimaki, T., Koenig, W., Jamshidi, Y., Siest, S., Abbasi, A., Uitterlinden, A. G., Abdollahi, M., Schnabel, R., Schick, U. M., Nolte, I. M., Kraja, A., Hsu, Y. H., Tylee, D. S., Zwicker, A., Uher, R., Davey-Smith, G., Morrison, A. C., Hicks, A., van Duijn, C. M., Ward-Caviness, C., Boerwinkle, E., Rotter, J., Rice, K., Lange, L., Perola, M., de Geus, E., Morris, A. P., Makela, K. M., Stacey, D., Eriksson, J., Frayling, T. M., & Slagboom, E. P. (2018). Genome analyses of >200,000 individuals identify 58 loci for chronic inflammation and highlight pathways that link inflammation and complex disorders. American Journal of Human Genetics, 103(5), 691–706.

    PubMed  PubMed Central  Google Scholar 

  • Lintas, C. (2019). Linking genetics to epigenetics: The role of folate and folate-related pathways in neurodevelopmental disorders. Clinical Genetics, 95(2), 241–252.

    PubMed  Google Scholar 

  • Lovallo, W. R. (2006). The hypothalamic-pituitary-adrenocortical axis in addiction. International Journal of Psychophysiology, 59(3), 193–194.

    PubMed  PubMed Central  Google Scholar 

  • Lowe, J., Qeadan, F., Leeman, L., Shrestha, S., Stephen, J. M., & Bakhireva, L. N. (2017). The effect of prenatal substance use and maternal contingent responsiveness on infant affect. Early Human Development, 115, 51–59.

    PubMed  PubMed Central  Google Scholar 

  • Lupien, S. J., McEwen, B. S., Gunnar, M. R., & Heim, C. (2009). Effects of stress throughout the lifespan on the brain, behaviour and cognition. Nature Reviews Neuroscience, 10(6), 434–445.

    PubMed  Google Scholar 

  • Lynch, S., Pfeiffer, C. M., Georgieff, M. K., Brittenham, G., Fairweather-Tait, S., Hurrell, R. F., et al. (2018). Biomarkers of nutrition for development (BOND)—Iron review. The Journal of Nutrition, 148(suppl_1), 1001S–1067S.

    PubMed  PubMed Central  Google Scholar 

  • Manns-James, L., & Neal-Barnett, A. (2019). Development of a culturally informed protocol for hair cortisol sampling in Black women. Public Health Nursing, 36(6), 872–879.

    PubMed  Google Scholar 

  • Martinez, M. (1992). Tissue levels of polyunsaturated fatty acids during early human development. Journal of Pediatrics, 120(4 Pt 2), S129–S138.

    Google Scholar 

  • Mastorakos, G., & Ilias, I. (2003). Maternal and fetal hypothalamic-pituitary-adrenal axes during pregnancy and postpartum. The Annals of the New York Academy of Sciences, 997, 136–149.

    PubMed  Google Scholar 

  • McEwen, B. S., & Gianaros, P. J. (2010). Central role of the brain in stress and adaptation: Links to socioeconomic status, health, and disease. The Annals of the New York Academy of Sciences, 1186, 190–222.

    PubMed  Google Scholar 

  • McLachlan, K., Rasmussen, C., Oberlander, T. F., Loock, C., Pei, J., Andrew, G., Reynolds, J., & Weinberg, J. (2016). Dysregulation of the cortisol diurnal rhythm following prenatal alcohol exposure and early life adversity. Alcohol, 53, 9–18.

    PubMed  PubMed Central  Google Scholar 

  • Mills, M. C., & Rahal, C. (2019). A scientometric review of genome-wide association studies. Commun Biol, 2, 9. https://doi.org/10.1038/s42003-018-0261-x.

    Article  PubMed  PubMed Central  Google Scholar 

  • Milman, N., & Kirchhoff, M. (1996). Relationship between serum ferritin, alcohol intake, and social status in 2235 Danish men and women. Annals of Hematology, 72(3), 145–151.

    PubMed  Google Scholar 

  • Msall, M. E., Bier, J. A., LaGasse, L., Tremont, M., & Lester, B. (1998). The vulnerable preschool child: The impact of biomedical and social risks on neurodevelopmental function. Seminars in Pediatric Neurology, 5(1), 52–61.

    PubMed  Google Scholar 

  • National Institutes of Health (2016). Environmental Influences on Child Health Outcomes (ECHO) program. https://www.nih.gov/research-training/environmental-influences-child-health-outcomes-echo-program. Accessed 8/11/2020.

  • Nessel, I., Khashu, M., & Dyall, S. C. (2019). The effects of storage conditions on long-chain polyunsaturated fatty acids, lipid mediators, and antioxidants in donor human milk—A review. Prostaglandins, Leukotrienes and Essential Fatty Acids, 149, 8–17.

    Google Scholar 

  • Obeid, R., Munz, W., Jäger, M., Schmidt, W., & Herrmann, W. (2005). Biochemical indexes of the B vitamins in cord serum are predicted by maternal B vitamin status. The American Journal of Clinical Nutrition, 82(1), 133–139.

    PubMed  Google Scholar 

  • Ornoy, A. (2003). The impact of intrauterine exposure versus postnatal environment in neurodevelopmental toxicity: Long-term neurobehavioral studies in children at risk for developmental disorders. Toxicology Letters, 140, 171–181.

    PubMed  Google Scholar 

  • Palmeri, A., Pichini, S., Pacifici, R., Zuccaro, P., & Lopez, A. (2000). Drugs in nails: Physiology, pharmacokinetics and forensic toxicology. Clinical Pharmacokinetics, 38(2), 95–110.

    PubMed  Google Scholar 

  • Paskett, E. D., Reeves, K. W., McLaughlin, J. M., Katz, M. L., McAlearney, A. S., Ruffin, M. T., Halbert, C. H., Merete, C., Davis, F., & Gehlert, S. (2008). Recruitment of minority and underserved populations in the United States: The centers for population health and health disparities experience. Contemporary Clinical Trials, 29(6), 847–861.

    PubMed  PubMed Central  Google Scholar 

  • Paul, S. E., Hatoum, A. S., Fine, J. D., Johnson, E. C., Hansen, I., Karcher, N. R., et al. (2019). Prenatal cannabis exposure and childhood outcomes: Results from the ABCD study. medRxiv https://www.medrxiv.org/content/medrxiv/early/2019/12/19/2019.12.18.19015164.full.pdf.

  • Pellizzari, E. D., Woodruff, T. J., Boyles, R. R., Kannan, K., Beamer, P. I., Buckley, J. P., Wang, A., Zhu, Y., Bennett, D. H., & (Environmental influences on Child Health Outcomes). (2019). Identifying and prioritizing chemicals with uncertain burden of exposure: Opportunities for biomonitoring and health-related research. Environmental Health Perspectives, 127(12), 126001.

    PubMed  PubMed Central  Google Scholar 

  • Pessah, I. N., Lein, P. J., Seegal, R. F., & Sagiv, S. K. (2019). Neurotoxicity of polychlorinated biphenyls and related organohalogens. Acta Neuropathologica, 138(3), 363–387.

    PubMed  PubMed Central  Google Scholar 

  • Petrick, L. M., Arora, M., & Niedzwiecki, M. M. (2020). Minimally Invasive Biospecimen Collection for Exposome Research in Children’s Health. Curr Environ Health Rep, 7(3), 198–210. https://doi.org/10.1007/s40572-020-00277-2

  • Polderman, T. J., Benyamin, B., de Leeuw, C. A., Sullivan, P. F., van Bochoven, A., Visscher, P. M., & Posthuma, D. (2015). Meta-analysis of the heritability of human traits based on fifty years of twin studies. Nat Genet, 47(7), 702–9. https://doi.org/10.1038/ng.3285.

    Article  PubMed  Google Scholar 

  • Popova, S., Lange, S., Shield, K., Mihic, A., Chudley, A. E., Mukherjee, R. A. S., Bekmuradov, D., & Rehm, J. (2016). Comorbidity of fetal alcohol spectrum disorder: A systematic review and meta-analysis. Lancet, 387(10022), 978–987.

    PubMed  Google Scholar 

  • Robinson, K. A., Dennison, C. R., Wayman, D. M., Pronovost, P. J., & Needham, D. M. (2007). Systematic review identifies number of strategies important for retaining study participants. Journal of Clinical Epidemiology, 60(8), 757–765.

    PubMed  PubMed Central  Google Scholar 

  • Rosen, L. J., Tillinger, E., Guttman, N., Rosenblat, S., Zucker, D. M., Stillman, F., & Myers, V. (2015). Parental receptivity to child biomarker testing for tobacco smoke exposure: A qualitative study. Patient Education and Counseling, 98(11), 1439–1445.

    PubMed  Google Scholar 

  • Sandman, C. A., Glynn, L., Schetter, C. D., Wadhwa, P., Garite, T., Chicz-DeMet, A., & Hobel, C. (2006). Elevated maternal cortisol early in pregnancy predicts third trimester levels of placental corticotropin releasing hormone (CRH): Priming the placental clock. Peptides, 27(6), 1457–1463.

    PubMed  Google Scholar 

  • Sandtorv, L. B., Hysing, M., Rognlid, M., Nilsen, S. A., & Elgen, I. B. (2017). Mental health in school-aged children prenatally exposed to alcohol and other substances. Substance Abuse, 11, 1178221817718160.

    PubMed  Google Scholar 

  • Santos, L. (2008). Genetic research in native communities. Progress in Community Health Partnerships, 2(4), 321–327.

    PubMed  PubMed Central  Google Scholar 

  • Sarangarm, P., Young, B., Rayburn, W., Jaiswal, P., Dodd, M., Phelan, S., & Bakhireva, L. (2012). Agreement between self-report and prescription data in medical records for pregnant women. Birth Defects Research Part A: Clinical and Molecular Teratology, 94(3), 153–161.

    PubMed  Google Scholar 

  • Shaw, L. P., Bassam, H., Barnes, C. P., Walker, A. S., Klein, N., & Balloux, F. (2019). Modelling microbiome recovery after antibiotics using a stability landscape framework. The ISME Journal, 13(7), 1845–1856.

    PubMed  PubMed Central  Google Scholar 

  • Sithisarn, T., Granger, D. T., & Bada, H. S. (2012). Consequences of prenatal substance use. International Journal of Adolescent Medicine and Health, 24(2), 105–112.

    PubMed  Google Scholar 

  • Smith, D. K., Johnson, A. B., Pears, K. C., Fisher, P. A., & DeGarmo, D. S. (2007). Child maltreatment and foster care: Unpacking the effects of prenatal and postnatal parental substance use. Child Maltreatment, 12(2), 150–160.

    PubMed  Google Scholar 

  • Spratt, E. G., Marsh, C., Wahlquist, A. E., Papa, C. E., Nietert, P. J., Brady, K. T., Herbert, T. L., & Wagner, C. (2016). Biologic effects of stress and bonding in mother–infant pairs. The International Journal of Psychiatry in Medicine, 51(3), 246–257.

    PubMed  Google Scholar 

  • Stragierowicz, J., Mikołajewska, K., Zawadzka-Stolarz, M., Polańska, K., & Ligocka, D. (2013). Estimation of cutoff values of cotinine in urine and saliva for pregnant women in Poland. Biomedical Research International, 2013, 386784.

    Google Scholar 

  • Strickland, J. C., Havens, J. R., & Stoops, W. W. (2019). A nationally representative analysis of "twin epidemics": Rising rates of methamphetamine use among persons who use opioids. Drug and Alcohol Dependence, 204, 107592.

    PubMed  PubMed Central  Google Scholar 

  • Sullivan, P. F., Agrawal, A., Bulik, C. M., Andreassen, O. A., Børglum, A. D., Breen, G., Cichon, S., Edenberg, H. J., Faraone, S. V., Gelernter, J., Mathews, C. A., Nievergelt, C. M., Smoller, J. W., O’Donovan, M. C., & for the Psychiatric Genomics Consortium. (2018). Psychiatric genomics: An update and an agenda. The American Journal of Psychiatry, 175(1), 15–27.

    PubMed  Google Scholar 

  • Szutorisz, H., & Hurd, Y. L. (2018). High times for cannabis: Epigenetic imprint and its legacy on brain and behavior. Neuroscience and Biobehavioral Reviews, 85, 93–101.

    PubMed  Google Scholar 

  • Thompson, P. M., Jahanshad, N., Ching, C. R. K., Salminen, L. E., Thomopoulos, S. I., Bright, J., et al. (2020). ENIGMA and global neuroscience: A decade of large-scale studies of the brain in health and disease across more than 40 countries. Translational Psychiatry, 10(1), 100.

    PubMed  PubMed Central  Google Scholar 

  • Torres, S., Merino, C., Paton, B., Correig, X., & Ramírez, N. (2018). Biomarkers of exposure to secondhand and thirdhand tobacco smoke: Recent advances and future perspectives. International Journal of Environmental Research and Public Health, 15(12), 2693.

  • Tsigos, C., Kyrou, I., Kassi, E., & Chrousos, G. P. (2000). Stress, endocrine physiology and pathophysiology. In K. R. Feingold, B. Anawalt, A. Boyce, G. Chrousos, K. Dungan, A. Grossman, et al. (Eds.), Endotext. South Dartmouth (MA): MDText.com, Inc..

    Google Scholar 

  • Turecki, G., & Meaney, M. J. (2016). Effects of the social environment and stress on glucocorticoid receptor gene methylation: A systematic review. Biological Psychiatry, 79(2), 87–96.

    PubMed  Google Scholar 

  • Ulrich-Lai, Y. M., & Herman, J. P. (2009). Neural regulation of endocrine and autonomic stress responses. Nature Reviews. Neuroscience, 10(6), 397–409.

    PubMed  PubMed Central  Google Scholar 

  • van den Berg, M., Kypke, K., Kotz, A., Tritscher, A., Lee, S. Y., Magulova, K., Fiedler, H., & Malisch, R. (2017). WHO/UNEP global surveys of PCDDs, PCDFs, PCBs and DDTs in human milk and benefit–risk evaluation of breastfeeding. Archives of Toxicology, 91(1), 83–96.

    PubMed  Google Scholar 

  • Volkow, N. D., Han, B., Compton, W. M., & McCance-Katz, E. F. (2019). Self-reported medical and nonmedical cannabis use among pregnant women in the United States. Journal of the American Medical Association, 322(2), 167–169.

    PubMed  Google Scholar 

  • Vuong, A. M., Yolton, K., Dietrich, K. N., Braun, J. M., Lanphear, B. P., & Chen, A. (2018). Exposure to polybrominated diphenyl ethers (PBDEs) and child behavior: Current findings and future directions. Hormones and Behavior, 101, 94–104.

    PubMed  Google Scholar 

  • Vuong, H. E., & Hsiao, E. Y. (2017). Emerging roles for the gut microbiome in autism spectrum disorder. Biological Psychiatry, 81(5), 411–423.

    PubMed  Google Scholar 

  • Wachman, E. M., & Farrer, L. A. (2019). The genetics and epigenetics of neonatal abstinence syndrome. Seminars in Fetal and Neonatal Medicine, 24(2), 105–110.

    PubMed  Google Scholar 

  • Wahono, N. A. (2018). Deciduous teeth as a tool to record early life exposure of zinc nutrition (Doctoral dissertation, Newcastle University). https://theses.ncl.ac.uk/jspui/bitstream/10443/4290/1/Wahono,%20N.A.%20%202018.pdf.

  • Weaver, I. C., Cervoni, N., Champagne, F. A., D'Alessio, A. C., Sharma, S., Seckl, J. R., et al. (2004). Epigenetic programming by maternal behavior. Nature Neuroscience, 7(8), 847–854.

    PubMed  Google Scholar 

  • Wu, F., Marin, S. J., & McMillin, G. A. (2017). Stability of 21 cocaine, opioid and benzodiazepine drug analytes in spiked meconium at three temperatures. Joural of Analytical Toxicology, 41(3), 196–204.

    Google Scholar 

  • Yao, R. A., Akinrinade, O., Chaix, M., & Mital, S. (2020). Quality of whole genome sequencing from blood versus saliva derived DNA in cardiac patients. BMC Medical Genomics, 13(1), 11.

    PubMed  PubMed Central  Google Scholar 

  • Yuan, N., Chen, Y., Xia, Y., Dai, J., & Liu, C. (2019). Inflammation-related biomarkers in major psychiatric disorders: A cross-disorder assessment of reproducibility and specificity in 43 meta-analyses. Translational Psychiatry, 9(1), 233.

    PubMed  PubMed Central  Google Scholar 

  • Zelner, I., Hutson, J. R., Kapur, B. M., Feig, D. S., & Koren, G. (2012). False-positive meconium test results for fatty acid ethyl esters secondary to delayed sample collection. Alcoholism, Clinical and Experimental Research, 36(9), 1497–1506.

    PubMed  Google Scholar 

  • Zhao, Y., Liu, P., Wang, J., Xiao, X., Meng, X., & Zhang, Y. (2016). Umbilical cord blood PBDEs concentrations are associated with placental DNA methylation. Environment International, 97, 1–6.

    PubMed  Google Scholar 

  • Zhou, S., Rosenthal, D. G., Sherman, S., Zelikoff, J., Gordon, T., & Weitzman, M. (2014). Physical, behavioral, and cognitive effects of prenatal tobacco and postnatal secondhand smoke exposure. Current Problems in Pediatric and Adolescent Health Care, 44(8), 219–241.

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgments

We wish to acknowledge the contributions of the biospecimen working group for the HEALthy Brain and Child Development study for their thoughtful contributions. These members include Arpana Agrawal (Washington University), Gretchen Bandoli (University of California San Diego), Veerle Bergink (Mt. Sinai), Christopher Coe (University of Wisconsin), Jessica Coker (University of Arkansas), Doug Dean (University of Wisconsin), Suzanne Devkota (Cedars Sinai Medical Center), Amy Elliott (Avera), Dani Fallin (Johns Hopkins University), Michael Georgieff (University of Minnesota), Michael Goran (Children’s Hospital of Los Angeles), Karen Grewen (University of North Carolina), Matthew Gurka (University of Florida), Sudha Iyengar (Case Western Reserve University), Suenna Massey (Northwestern University), Debra Mathews (Johns Hopkins University), Kay Roussos-Ross (University of Florida), Elinor Sullivan (Oregon Health Sciences University), and T. Kent Teague (University of Oklahoma). We also wish to acknowledge the thoughtful and meaningful contributions of the team supporting this effort at the National Institutes of Health, including Drs. Michelle Freund, Chloe Jordan, Yuxia Cui, Minki Chatterji, Julia Zehr, Kimberley LeBlanc, and Katia Delrahim Howlett.

Funding

This work was supported by the National Institute on Drug Abuse (NIDA) research grant numbers R34 R34DA050343 (JMC), R34 DA050272 (RB), R34DA050292 (SBJ), and R34 DA050237 (LNB).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Julie M. Croff.

Ethics declarations

Disclaimer

The views and opinions expressed in this manuscript are those of the authors only and do not necessarily represent the views, official policy, or position of the US Department of Health and Human Services or any of its affiliated institutions or agencies.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Croff, J.M., Bogdan, R., Johnson, S.B. et al. Early Environmental Exposures and Contaminants: a Design Framework for Biospecimen Collection and Analysis for a Prospective National Birth Cohort. ADV RES SCI 1, 269–283 (2020). https://doi.org/10.1007/s42844-020-00024-4

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s42844-020-00024-4

Keywords

Navigation