Skip to main content
Open access
Research
23 June 2022

Associations of Pre- and Postnatal Air Pollution Exposures with Child Behavioral Problems and Cognitive Performance: A U.S. Multi-Cohort Study

Publication: Environmental Health Perspectives
Volume 130, Issue 6
CID: 067008

Abstract

Background:

Population studies support the adverse associations of air pollution exposures with child behavioral functioning and cognitive performance, but few studies have used spatiotemporally resolved pollutant assessments.

Objectives:

We investigated these associations using more refined exposure assessments in 1,967 mother–child dyads from three U.S. pregnancy cohorts in six cities in the ECHO-PATHWAYS Consortium.

Methods:

Pre- and postnatal nitrogen dioxide ( NO 2 ) and particulate matter (PM) 2.5 μ m in aerodynamic diameter ( PM 2.5 ) exposures were derived from an advanced spatiotemporal model. Child behavior was reported as Total Problems raw score using the Child Behavior Checklist at age 4–6 y. Child cognition was assessed using cohort-specific cognitive performance scales and quantified as the Full-Scale Intelligence Quotient (IQ). We fitted multivariate linear regression models that were adjusted for sociodemographic, behavioral, and psychological factors to estimate associations per 2-unit increase in pollutant in each exposure window and examined modification by child sex. Identified critical windows were further verified by distributed lag models (DLMs).

Results:

Mean NO 2 and PM 2.5 ranged from 8.4  to  9.0  ppb and 8.4  to  9.1 μ g / m 3 , respectively, across pre- and postnatal windows. Average child Total Problems score and IQ were 22.7 [standard deviation (SD): 18.5] and 102.6 (SD: 15.3), respectively. Children with higher prenatal NO 2 exposures were likely to have more behavioral problems [ β : 1.24; 95% confidence interval (CI): 0.39, 2.08; per 2  ppb NO 2 ], particularly NO 2 in the first and second trimester. Each 2 - μ g / m 3 increase in PM 2.5 at age 2–4 y was associated with a 3.59 unit (95% CI: 0.35, 6.84) higher Total Problems score and a 2.63 point (95% CI: 5.08 , 0.17 ) lower IQ. The associations between PM 2.5 and Total Problems score were generally stronger in girls. Most predefined windows identified were not confirmed by DLMs.

Discussion:

Our study extends earlier findings that have raised concerns about impaired behavioral functioning and cognitive performance in children exposed to NO 2 and PM 2.5 in utero and in early life. https://doi.org/10.1289/EHP10248

Introduction

Early brain morphology in humans begins in the third week post conception and rapidly develops by midgestation.13 Ongoing structural change and functional development continue for an extended period postnatally until early adulthood.1,4,5 Subtle disturbances in early life may interfere with the normal trajectory of brain development and cause subsequent functional impairment.2 Children with behavioral and cognitive impairment early in life may have persistent problems, including increased risk of substance abuse, violent behavior, and depression in adolescence and/or adulthood,6 as well as diminished academic performance and economic productivity over their life span.7,8 Therefore, identifying modifiable factors on which to intervene is a research priority. In recent decades, growing evidence has demonstrated the human neurodevelopmental toxicity of common air pollutants, including nitrogen oxides ( NO 2 ), particulate matter (PM) 2.5 μ m in aerodynamic diameter ( PM 2.5 ), and polycyclic aromatic hydrocarbons (PAHs), on the central nervous system (CNS) with subsequent behavioral and cognitive impacts.911
Air pollutants can invade deep in the lungs, trigger oxidative stress, and induce systemic inflammation in pregnant women.1214 Circulating markers pass the maternal–fetal blood barrier and promote chronic inflammation and neurodegeneration in the fetus,1517 with evidence of longer-term impact on offspring neurodevelopmental outcomes.1820 Postnatal air pollution exposures may affect children’s CNS more directly. Besides penetrating into the lungs, inhaled pollutants may also translocate along the olfactory nerve into the olfactory bulb, promote diffusion of oxidative stress and inflammatory markers across the impaired blood brain barrier, and induce microglial activation on entering the CNS.2124 Previous population studies in the United States,2528 Europe,2931 and Asia3234 have consistently linked air pollution exposures in both pre- and postnatal windows to poorer neurodevelopmental outcomes during early to late childhood. Nevertheless, exposure data at a small scale are scarce and available in only a few studies,28,32,3538 four of which estimated trimester specific associations.28,32,36,37 In these previous studies, various pollutants (mainly NO 2 and PM) were estimated from a number of exposure assessment methods, including conventional geographic information system-based methods (e.g., land use regression), direct measurements from residential ambient monitors, and biomarkers of exposure.10 The use of spatiotemporally resolved air pollution assessment across different developmental windows in a multiregion sample is limited. Other methodological limitations in prior studies include inadequate assessment of confounding, failure to consider outcomes across neurodevelopmental domains, and inability to disentangle the associations from distinct exposure windows due to limited statistical power and/or air pollution predictions with a low temporal resolution.
In this study, we further the current body of literature and our own recent analyses27,39 of NO 2 and PM 10 with child behavioral problems and intelligence quotient (IQ). Specifically, here we expand the study population to include three pregnancy cohorts in the United States, use spatiotemporally resolved predictions of NO 2 and PM 2.5 , and evaluate several sensitive developmental periods in both pre- and postnatal windows. We hypothesized that children with higher pre- and/or postnatal exposures to NO 2 and PM 2.5 would have more behavioral problems and a lower IQ. In addition, research has identified several sex differences in neurodevelopment, including morphological, physiological, and chemical differences.40 Several lines of evidence suggest that the air pollution–child neurodevelopment association may be sex-specific, possibly with a more pronounced relationship in boys.25,30,35,4143 However, heterogeneities in study design of previous studies, including varied exposure and outcome assessments, have hampered the ability to draw firm conclusions regarding differential vulnerability by sex. Hence, we also examined whether the associations of interest differed by child sex in the current study.

Methods

Study Population

As part of the Environmental Influences on Child Health Outcomes (ECHO) study, we aimed to maximize power in the current analysis to estimate effects from air pollution exposures on child behavioral and cognitive outcomes and explore nuanced research questions of effect modification by child sex by pooling all three U.S. pregnancy cohorts that compose the ECHO-PATHWAYS Consortium: the Conditions Affecting Neurocognitive Development and Learning in Early Childhood (CANDLE) study, the Infant Development and Environment Study (TIDES), and the Global Alliance to Prevent Prematurity and Stillbirth (GAPPS). Based in Shelby County, Tennessee, the CANDLE study initially aimed to identify risk factors for child neurodevelopment.44 Women were considered eligible if they were 16–40 y old, had medically low-risk singleton pregnancies, and planned to deliver at a participating study hospital. From 2006 to 2011, 1,503 women were recruited in their second trimester from either the general community or affiliated medical group clinics. More than three quarters of the enrolled participants ( n = 1,143 ) completed a clinic visit when the resulting children were age 4–6 y. The TIDES study was designed to examine the impacts of exposure to endocrine disrupting chemicals, notably phthalates, on child health and development.45 From 2010 to 2012, recruitment commenced in academic medical centers in four cities: San Francisco, California; Rochester, New York; Minneapolis, Minnesota; and Seattle, Washington. Pregnant women in their first trimester were considered eligible if they were age 18 y or older, were English-speaking, had singleton pregnancies without any serious threat, and planned to deliver at a participating study hospital. There were 749 women who were retained in the study throughout the pregnancy and delivered a live birth, and 551 mother–child dyads completed the 6-y visit. Last, the GAPPS study was established to inform evidence-based treatments and interventions to reduce preterm birth and stillbirth through development of a biorepository (www.gapps.org). Families who had participated in the GAPPS study and met eligibility criteria (consented to contact for future study; had prenatal questionnaire data and biospecimens available; child age was eligible for the 4–6-y visit) were invited to participate in the ECHO-PATHWAYS Consortium. From July 2017 to April 2020, 439 mother–child dyads from Seattle and Yakima, Washington, were enrolled and completed the follow-up visit at child age 4–6 y.
The current study included 1,967 CANDLE, TIDES, and GAPPS children who completed behavioral and cognitive assessments at clinical visits at 4–6 y of age and had valid residential addresses in the pre- and/or postnatal windows reported by parents. Each woman provided informed consent upon enrollment in original cohorts. This analysis uses previously collected data from the three cohorts and was approved by the Human Subjects Division at the University of Washington.

Child Cognitive and Behavioral Measurements

Child behaviors were assessed using the Child Behavior Checklist (CBCL), administered at a visit that occurred at age 4–6 y. The CBCL involves caregiver reporting of a wide range of emotional and behavior problems in children and is broadly used in both research and clinical settings.46,47 One of two CBCL versions were administered, depending on the child’s age: the CBCL preschool form (ages 1.5–5 y)48 or the CBCL school-age form (ages 6–18 y).49 All CANDLE participants completed the preschool form, all TIDES participants completed the school-age forms, and GAPPS families completed a mix of both forms. The preschool form includes report of the frequency of 99 child behaviors in the past 2 months, whereas the school-age form includes 112 behaviors in the past 6 months. Caregivers rate these items on a scale of Not True (0), Somewhat or Sometimes True (1), to Very True or Often True (2). Although additional behaviors appropriate for children up to age 18 y are added in the school-age form, given that the children in our study were essentially in the same developmental stage, caregivers reported similar types and counts of behaviors across forms. Therefore, we combined the data collected by two CBCL forms. Because the standardized t-scores estimated from these two CBCL forms differ in whether child sex was adjusted,50 we computed the raw score for the Total Problems scale as the primary outcome and further calculated the standardized z-score using all ECHO-PATHWAYS participants with behavioral problems measured by either CBCL form as the reference to verify our findings.
Child cognitive performance was examined at the same visit as the behavior measurement. The IQs of the CANDLE, TIDES, and GAPPS children were assessed using the Stanford-Binet Intelligence Scales, Fifth Edition (SB-5),51,52 the abbreviated five-subtest version of the Wechsler Intelligence Scale for Children, Fifth Edition (WISC-V),53,54 and the Wechsler Preschool & Primary Scale of Intelligence, Fourth Edition (WPPSI-IV, age 4:0–7:7 version),55,56 respectively. The three IQ batteries are examiner-administered, highly reliable, and valid measures of intellectual and cognitive abilities in childhood. All examiners were trained on the administration and scoring by licensed psychologists. They participated in didactic instruction and guided practice, interrater reliability exercises, as well as weekly supervision by psychologists post training. Considering that Full-Scale IQ is less frequently included in large population studies due to its heavy burden on examiners and participants, we aimed to maximize use of these data as others have before us.5759 In CANDLE, full-scale IQ was derived from the 10 subtests in the SB-5 addressing five cognitive factors with verbal and nonverbal tests for each factor: knowledge, fluid reasoning, quantitative reasoning, visual-spatial processing, and working memory. In TIDES, Full-Scale IQ was estimated using the Tellegen and Briggs formula,60 incorporating five WISC-V domains—verbal comprehension, visual spatial, fluid reasoning, working memory, and processing speed. The calculation of Full-Scale IQ from the WPPSI-IV in GAPPS included scores in the five cognitive domains, similar to the five WISC-V domains in TIDES. Although the specific tests used to capture different domains of cognitive performance may vary across the IQ instruments, full-scale IQ provides a standardized metric of overall performance across all the subtests, with a high correlation across instruments.61

Air Pollution Assessments

Detailed residential addresses were collected from CANDLE participants at enrollment and updated at each subsequent point of contact. The availability of address data varied by site in the TIDES study: all participants reported residential addresses at enrollment, at the age 4 y visit, and at the age 6 y visit; participants in Rochester and San Francisco were contacted for one more update between enrollment and the age 4 y visit. GAPPS participants were asked to provide comprehensive address histories at the age 4–6 y visit retrospectively from enrollment to present. Point-based NO 2 and PM 2.5 exposures were estimated from a spatiotemporal model on a 2-wk scale.62 This model used monitoring data from regulatory networks, further enhanced with air pollution measurements from intensive research cohort-specific monitors. A geographic information system was used to identify covariates representing land use characteristics that could reflect spatial variability in air pollution distributions, and the dimension-reduced regression covariates were obtained using partial least squares from more than 400 geographic variables. The space–time features of pollution concentrations were decomposed into spatially varying long-term averages, spatially varying seasonal and long-term trends, and spatially correlated but temporally independent residuals, and these components were fitted jointly in a likelihood-based spatiotemporal extension of universal kriging. We estimated biweekly NO 2 and PM 2.5 predictions from region-specific models (three regions for the NO 2 models and nine regions for the PM 2.5 models), and averaged the exposure concentrations over each trimester, the whole pregnancy, and the two postnatal windows from childbirth to 2 y old and from 2 to 4 y old. The 2–4-y PM 2.5 estimates were missing in 150 participants whose 4-y-old birthday was beyond the prediction window of the spatiotemporal model (30 December 1998 to 4 July 2017). Moving was accounted for by calculating the time-weighted averages of NO 2 and PM 2.5 in the relevant windows based on the reported move date. For families who moved between two points of contact and did not report a move date, we estimated the move date as the midpoint of the two contact dates. We refer readers to a recent paper by Kirwa et al. (2021)63 for a thorough discussion of different air pollution prediction models.

Covariates

Several indicators for maternal, child, and family characteristics, including multilevel social determinants, were harmonized across cohorts. Maternal characteristics included age at enrollment; region- and inflation-adjusted household annual income64; household members (2–3 vs. 4 vs. 5 vs. 6); education level ( < high school vs. high school/Graduate Equivalency Diploma vs. college/technical school vs. graduate or professional degree); marital status (married/living as married vs. single/living as single); pregnancy smoking (smoker vs. nonsmoker); pregnancy alcohol consumption (ever vs. never); pregnancy vitamin supplement intake (ever vs. never); prepregnancy body mass index (BMI); IQ measured by Wechsler Abbreviated Scale of Intelligence [the composite score of four subtests (Vocabulary, Similarities, Block Design, and Matrix Reasoning) from the first edition65 in CANDLE, the composite score of two subtests (Vocabulary and Matrix Reasoning) from the second edition66,67 in TIDES and GAPPS]; depression, assessed at the visit when child outcome assessments were taken, by either the Center for Epidemiologic Studies Depression Scale (CES-D)68 or the Patient-Reported Outcomes Measurement Information System (PROMIS)69 and quantified as t-scores; and breastfeeding practice (ever vs. never). Child characteristics included age at cognitive and behavioral assessments; sex (male vs. female); birth order (first born vs. not first born); birth year; and secondhand smoking exposure (anyone living in the child’s home smoked vs. no one smoked). The indices in two of the three domains of the Childhood Opportunity Index (COI) were calculated based on the residential address history in pre- and postnatal windows: A larger index in the social and economic subscale reflected higher neighborhood-level socioeconomic status (SES), and a larger index in the educational subscale indicated better early childhood education quality.70 We also included parent-reported child race (White vs. Black, vs. others) as a confounder, given that race is not only a proxy for racial residential segregation, which directly associates with air pollution exposures, but also a strong predictor of socioeconomic position that results in health disparities.71 American Indian/Alaska Native, Asian, and Native Hawaiian/other Pacific Islanders were grouped together to improve statistical power and enhance the data harmonizability across cohorts.

Statistical Analyses

We conducted descriptive analyses in the entire sample and by cohort to summarize the characteristics of the participants, the air pollution exposures, and the child cognitive and behavioral assessments. Linear regressions with robust standard errors were performed to estimate the associations of individual exposures ( PM 2.5 or NO 2 ) in each window with child Total Problems score and IQ based on observations with complete data pooled from three cohorts. To enable comparisons with studies with relatively low air pollution levels and variabilities, effect estimates were rescaled to a 2-unit incremental increase, which approximates the interquartile ranges (IQRs) for long-term exposures in the six study sites. Based on substantive knowledge from existing literature, we developed a staged adjustment approach comprising three models, allowing us to explore the influence of increasing levels of adjustment on results. We further created a directed acyclic graph (DAG; Figure S1) to help visualize the relationships. Model 1 (the minimal model) controlled for basic demographics—child sex, child age at outcome assessments, and study site. An indicator of CBCL form was additionally included in the analysis of Total Problems score. We defined Model 2 as the primary model, which was further adjusted for major confounders or precision variables, including child race, maternal education, log-transformed region- and inflation-adjusted household income, household members, an interaction between household members and income to account for nonproportional financial needs of a household grow with additional members,72 marital status, maternal age at enrollment, birth order, pregnancy smoking, pregnancy alcohol consumption, maternal depression, maternal IQ, child secondhand smoking exposure, and COI subscales (economics and education). Adjustment for covariates that are only associated with the outcomes (i.e., precision variables) in a linear setting is desirable, because it will improve the precision of the effect estimates by reducing residual variance.73 Model 3, an extended model, included additional adjustments for four covariates that may be proxies for unmeasured confounders74: Prepregnancy BMI, prenatal vitamin supplement intake, and breastfeeding may serve as proxies for maternal preventative behaviors, and child year of birth may act as a surrogate for birth cohort effects.
In a secondary analysis, we introduced cross product terms of child sex and individual air pollution exposures in each window in separate primary models (i.e., Model 2) and estimated the interaction p-values as well as sex-specific associations. Several sensitivity analyses were performed to verify the robustness of our main findings. First, we implemented complete case analysis as the primary approach. Although the data for most covariates were nearly complete in the analytic data set, 16.2% of the women did not have IQ measurement, and more than half of the missingness in this covariate came from the GAPPS study. To address the issue of missing data in maternal IQ and other covariates, we began by verifying the assumption that the chance of being a complete case solely depends on the observed covariates, where data were missing at random (MAR).75 We constructed the Receiver Operating Characteristic (ROC) curve by computing the metrics of sensitivity and specificity at various threshold settings via 5-fold cross-validation76 and further estimated the area under the ROC curve—a measure indicating the probability of models with the observed covariates to correctly distinguish complete cases from noncomplete cases.77 The area under the ROC curve indicated that this MAR assumption was plausible, and both complete case analysis and multiple imputation would give unbiased results. We thus employed multiple imputation by chained equations (MICE) in the minimal, primary, and extended models.75,78 Each missing value was imputed 10 times with 100 iterations between each round of imputation using predictive mean matching.79
Second, to investigate the validity of combining data of behavioral outcomes measured by different CBCL forms, we replaced the raw Total Problems score with the standardized z -score. Third, considering that children born earlier than 34 wk are at higher risks of substantially lower IQ, attention, and executive function in comparison with those with a gestational week 37 and above,80 we restricted the sample to those born at gestational week 34 or later. Fourth, to investigate whether the associations of interest were confounded by the exposures in the other windows, we simultaneously included NO 2 or PM 2.5 estimates in all three trimesters in the primary models and further controlled for postnatal exposures. In addition, we mutually adjusted for exposures over the whole pregnancy and postnatal pollution estimates. Fifth, to further confirm the identified critical exposure windows, we fit fully adjusted constrained distributed lag models (DLM) of biweekly air pollution predictions with varied degrees of freedom (df) from 4 to 9. The prenatal windows were restricted to 38 wk, and those born prior to gestational week 36 were imputed with the measurements in the latest available biweekly window. The prenatal and postnatal biweekly exposures were modeled separately with adjustments for average exposures in the other period(s), considering the different biological mechanisms linking child neurodevelopment with air pollution exposures in utero and after birth. Sixth, to assess the linearity of the relationships between air pollution exposures and outcomes, we conducted generalized additive models with full adjustments in the overall sample, as well as in strata by child sex. Finally, to better understand the impacts of inherent heterogeneity across the cohorts and sites and potential modified confounding by site on the findings, we implemented three additional analyses: a leave-one-cohort-out analysis, a leave-one-site-out analysis, and a set of three models compared to the results of fixed effects models (i.e., the primary analysis), including fixed effects models with site–covariate interactions, mixed-effects models with a fixed intercept and random slopes by site, and mixed-effects models with site–covariate interactions. All analyses were conducted in R (version 3.6.5; R Development Core Team).

Results

Characteristics of the Study Participants

The inclusion of participants from enrollment to outcome assessment as well as the analytic sample sizes are illustrated in Figure 1. Among the total analytic sample of 1,967 mother–child dyads, the CANDLE, TIDES, and GAPPS studies contributed 53%, 27%, and 20% of the population, respectively (Table 1). Children were on average 5.2 y of age (SD: 1.0) at outcome measurements, and there was an approximately equal number of boys and girls. Nearly half (48%) were identified as White by their parents and 39% as Black. One-fifth of the children were living with at least one relative who smoked. Mothers had an average age of 28.5 y (SD: 6.0) at enrollment, 56% had a college degree and above, and 70% were married or living as married. The median region- and inflation-adjusted household income was $ 55,800  USD (IQR: 62,500). Maternal IQ was normally distributed, with a mean of 100 (SD: 17.4). The average t-score of CES-D or PROMIS was 48.5 (SD: 7.4), indicating a typical level of maternal depression on average. Compared across cohorts, the CANDLE cohort comprised a larger proportion of Black participants and low-income families, whereas the TIDES participants were relatively more educated, with a higher household income. The analytic sample was similar to the overall sample of participants at enrollment (Table S1). The distribution of child Total Problems score was slightly right skewed (Figure S2) with a median of 18 (IQR: 22), whereas child IQ was relatively normally distributed with a mean of 102.6 (SD: 15.3). Variations in the distribution of child outcomes by cohort were observed (Table 2).
Figure 1. Shown are the inclusion of the three U.S. pregnancy cohorts in the ECHO-PATHWAYS Consortium (the CANDLE, TIDES, and GAPPS study) from enrollment to the visit of child cognitive and behavioral assessments, as well as the analytic sample sizes remaining from the implementation of each exclusion criterion. Note: CANDLE, Conditions Affecting Neurocognitive Development and Learning in Early Childhood; ECHO, Environmental Influences on Child Health Outcomes; GAPPS, Global Alliance to Prevent Prematurity and Stillbirth; IQ, intelligence quotient; TIDES, The Infant Development and Environment Study.
Table 1 Characteristics of participants from the three U.S. pregnancy cohorts in the ECHO-PATHWAYS consortium (the CANDLE, TIDES, and GAPPS study) included in analysis.
    Cohort
Total CANDLE TIDES GAPPS
( n = 1,967 ) ( n = 1,035 ) ( n = 539 ) ( n = 393 )
Child characteristics
Child age at outcome assessment (y) 5.2 ( ± 1.0 ) 4.4 ( ± 0.6 ) 6.4 ( ± 0.4 ) 5.6 ( ± 0.7 )
 Missing 47 2 3 42
Child sex
 Male 963 (49%) 517 (50%) 251 (47%) 195 (50%)
 Female 1,003 (51%) 518 (50%) 287 (53%) 198 (50%)
 Missing 1 0 1 0
Child race
 White 917 (48%) 264 (26%) 354 (68%) 299 (78%)
 Black 752 (39%) 679 (67%) 64 (12%) 9 (2%)
 Others 246 (13%) 68 (7%) 101 (19%) 77 (20%)
 Missing 52 24 20 8
Birth order
 Not firstborn 1,134 (58%) 629 (61%) 244 (46%) 261 (67%)
 Firstborn 819 (42%) 406 (39%) 282 (54%) 131 (33%)
 Missing 14 0 13 1
Year of birth
 2007 85 (4%) 85 (8%) 0 (0%) 0 (0%)
 2008 201 (10%) 201 (19%) 0 (0%) 0 (0%)
 2009 246 (13%) 246 (24%) 0 (0%) 0 (0%)
 2010 283 (14%) 283 (27%) 0 (0%) 0 (0%)
 2011 468 (24%) 220 (21%) 197 (37%) 51 (13%)
 2012 417 (21%) 0 (0%) 304 (56%) 113 (29%)
 2013 175 (9%) 0 (0%) 38 (7%) 137 (35%)
 2014 86 (4%) 0 (0%) 0 (0%) 86 (22%)
 2015 6 (0%) 0 (0%) 0 (0%) 6 (2%)
Secondhand smoking exposure
 No 1,523 (80%) 704 (68%) 465 (96%) 354 (91%)
 Yes 378 (20%) 324 (32%) 19 (4%) 35 (9%)
 Missing 66 7 55 4
Maternal characteristics
Maternal age at enrollment (y) 28.5 ( ± 6.0 ) 26.3 ( ± 5.6 ) 31.0 ( ± 5.5 ) 31.0 ( ± 5.5 )
Missing 12 0 0 12
Region-, inflation-adjusted household income ( $ 1,000 ) 55.8 [62.5] 31.8 [50.9] 101.7 [107.7] 86.0 [67.1]
Missing 77 46 24 7
Household members
 2–3 410 (21%) 220 (21%) 124 (24%) 66 (17%)
 4 767 (40%) 361 (35%) 247 (48%) 159 (42%)
 5 435 (23%) 248 (24%) 95 (19%) 92 (24%)
6 311 (16%) 200 (19%) 47 (9%) 64 (17%)
 Missing 44 6 26 12
Maternal education
 Less than high school 167 (9%) 123 (12%) 32 (6%) 12 (3%)
 High school/GED 683 (35%) 495 (48%) 88 (16%) 100 (26%)
 College/technical school 644 (33%) 304 (29%) 169 (32%) 171 (45%)
 Graduate or Professional degree 458 (23%) 112 (11%) 246 (46%) 100 (26%)
 Missing 15 1 4 10
Marital status
 Married/living as married 1,365 (70%) 572 (55%) 456 (85%) 337 (88%)
 Single/living as single 589 (30%) 462 (45%) 82 (15%) 45 (12%)
 Missing 13 1 1 11
Pregnancy smoking
 Nonsmoker 1,828 (94%) 942 (91%) 514 (96%) 372 (97%)
 Smoker 122 (6%) 92 (9%) 19 (4%) 11 (3%)
 Missing 17 1 6 10
Pregnancy alcohol consumption
 No alcohol consumption 1,760 (90%) 947 (92%) 472 (88%) 341 (90%)
 Alcohol consumption 188 (10%) 87 (8%) 65 (12%) 36 (10%)
 Missing 19 1 2 16
Pregnancy supplement intake
 Never 260 (13%) 57 (6%) 181 (34%) 22 (6%)
 Ever 1,675 (87%) 958 (94%) 356 (66%) 361 (94%)
 Missing 32 20 2 10
Breastfeeding practice
 Never 443 (23%) 375 (36%) 46 (9%) 22 (6%)
 Ever 1,512 (77%) 658 (64%) 485 (91%) 369 (94%)
 Missing 12 2 8 2
Prepregnancy BMI ( kg / m 2 ) 27.1 ( ± 7.43 ) 28.0 ( ± 7.88 ) 25.5 ( ± 6.23 ) 27.0 ( ± 7.36 )
 Missing 45 3 14 28
Maternal IQa 100 ( ± 17.4 ) 94.6 ( ± 16.3 ) 109 ( ± 16.3 ) 108 ( ± 13.3 )
 Missing 328 12 111 205
Maternal depressionb 48.5 ( ± 7.4 ) 48.6 ( ± 6.9 ) 48.3 ( ± 8.2 ) 48.9 ( ± 7.8 )
 Missing 28 9 14 5
Child Opportunity Educational Index (Prenatal) 0.022 ( ± 0.073 ) 0.049 ( ± 0.064 ) 0.019 ( ± 0.072 ) 0.006 ( ± 0.067 )
 Missing 28 5 6 17
Child Opportunity Educational Index (0–2 y) 0.022 ( ± 0.073 ) 0.049 ( ± 0.063 ) 0.020 ( ± 0.073 ) 0.007 ( ± 0.066 )
 Missing 8 0 4 4
Child Opportunity Educational Index (2–4 y) 0.019 ( ± 0.074 ) 0.048 ( ± 0.065 ) 0.026 ( ± 0.070 ) 0.0028 ( ± 0.064 )
 Missing 16 2 9 5
Child Opportunity Economics Index (Prenatal) 0.045 ( ± 0.257 ) 0.133 ( ± 0.266 ) 0.057 ( ± 0.233 ) 0.051 ( ± 0.160 )
 Missing 28 5 6 17
Child Opportunity Economics Index (0–2 y) 0.042 ( ± 0.249 ) 0.131 ( ± 0.252 ) 0.060 ( ± 0.231 ) 0.054 ( ± 0.156 )
 Missing 8 0 4 4
Child Opportunity Economics Index (2–4 y) 0.034 ( ± 0.246 ) 0.126 ( ± 0.253 ) 0.073 ( ± 0.218 ) 0.063 ( ± 0.147 )
 Missing 16 2 9 5
Recruitment site
 Memphis 1,035 (53%) 1,035 (100%) 0 (0%) 0 (0%)
 San Francisco 135 (7%) 0 (0%) 135 (25%) 0 (0%)
 Minneapolis 151 (8%) 0 (0%) 151 (28%) 0 (0%)
 Rochester 135 (7%) 0 (0%) 135 (25%) 0 (0%)
 Seattle TIDES 118 (6%) 0 (0%) 118 (22%) 0 (0%)
 Seattle GAPPS 199 (10%) 0 (0%) 0 (0%) 199 (51%)
 Yakima 194 (10%) 0 (0%) 0 (0%) 194 (49%)
Note: Shown in the table are mean ( ± SD ), counts (percentage), and median (interquartile range); proportions were calculated in complete cases. BMI, body mass index; CANDLE, Conditions Affecting Neurocognitive Development and Learning in Early Childhood; ECHO, Environmental Influences on Child Health Outcomes; GAPPS, Global Alliance to Prevent Prematurity and Stillbirth; GED, general equivalency diploma; IQ, intelligence quotient; SD, standard deviation; TIDES, The Infant Development and Environment Study.
a
Maternal IQ was measured by Wechsler Abbreviated Scale of Intelligence [the composite score of four subtests (Vocabulary, Similarities, Block Design, and Matrix Reasoning) from the first edition in CANDLE, the composite score of two subtests (Vocabulary and Matrix Reasoning) from the second edition in TIDES and GAPPS].
b
Maternal depression was quantified using the t -scores at the visit of child outcome assessments by either the Center for Epidemiologic Studies Depression Scale (CES-D) or the Patient-Reported Outcomes Measurement Information System (PROMIS).
Table 2 Distributions of child total problems score and IQ in participants from the three U.S. pregnancy cohorts in the ECHO-PATHWAYS consortium (the CANDLE, TIDES, and GAPPS study).
Outcomes Cohort n Min 1st quartile Median Mean (SD) 3rd quartile Max
Total problem score Overall 1,895 0 9 18 22.66 (18.52) 31 132
 CANDLEa 1,005 0 10 19 23.72 (19.37) 33 132
 TIDESb 533 0 9 18 21.82 (17.13) 29 96
 GAPPS preschool form 262 0 9 15.5 20.96 (17.71) 28 110
 GAPPS school-age form 95 0 9 14 20.93 (18.6) 27.5 94
IQ Overall 1,810 40 93 104 102.57 (15.27) 113 149
 CANDLE 1,030 40 90 100 99.7 (14.85) 110 138
 TIDES 425 55 97 107 105.96 (16.37) 118 149
 GAPPS 355 52 99 108 106.86 (13.19) 115 136
Note: CANDLE, Conditions Affecting Neurocognitive Development and Learning in Early Childhood; ECHO, Environmental Influences on Child Health Outcomes; GAPPS, Global Alliance to Prevent Prematurity and Stillbirth; min, minimum; max, maximum; SD, standard deviation; TIDES, The Infant Development and Environment Study.
a
All CANDLE participants completed the preschool form.
b
All TIDES participants completed the school-age forms.

Air Pollution Exposures

NO 2 levels were relatively stable and did not show a clear pattern over time, with a mean ranging from 8.4 (SD: 3.8) to 9.0 (SD: 3.9) ppb in different pre- and postnatal windows (Table 3). The correlation between NO 2 in nonoverlapping windows was highest between overall pregnancy and age 0–2 y (Spearman correlation: 0.84) and lowest between the first and third trimester (Spearman correlation: 0.32) (Table S2). Prenatal PM 2.5 was marginally higher than the exposures in the two postnatal windows. The concentrations were 9.0 (SD: 2.3) μ g / m 3 , 8.8 (SD: 2.0) μ g / m 3 , and 8.4 (SD: 1.8) μ g / m 3 averaged over the pregnancy, 0–2 y, and 2–4 y, respectively. PM 2.5 aggregated in shorter windows had a greater variation than the exposures in the longer windows. There was a medium to high correlation of PM 2.5 across windows (Spearman correlation: 0.50–0.89), mainly driven by between-cohort correlations. The variation in PM 2.5 was lower than that in NO 2 , and PM 2.5 and NO 2 in the same period were moderately correlated (Spearman correlation: 0.01–0.47). The NO 2 concentrations in Seattle, San Francisco, and Minneapolis were greater than those in Memphis, Rochester, and Yakima (Figure S3). In contrast, Memphis had the highest PM 2.5 level, followed by San Francisco, Minneapolis, and Rochester. Seattle and Yakima in Washington state had the lowest PM 2.5 level.
Table 3 Distributions of NO 2 and PM 2.5 in each pre- and postnatal window in the overall analytic sample from the three U.S. pregnancy cohorts in the ECHO-PATHWAYS consortium (the CANDLE, TIDES, and GAPPS study).
Exposures Window n Min 1st quartile Median Mean (SD) 3rd quartile Max
NO 2 (ppb) 1st trimester 1,935 1.59 6.01 8.62 8.96 (3.92) 11.49 33.74
2nd trimester 1,934 1.44 5.66 7.97 8.45 (3.77) 10.82 29.15
3rd trimester 1,920 1.19 5.56 7.91 8.36 (3.82) 10.56 33.53
Overall pregnancy 1,932 1.74 6.36 8.35 8.62 (3.17) 10.65 27.29
0–2 y 1,894 1.63 6.79 8.78 8.71 (2.93) 10.53 26.37
2–4 y 1,894 1.61 6.46 8.68 8.59 (3.03) 10.50 25.93
PM 2.5 ( μ g / m 3 ) 1st trimester 1,935 1.82 7.33 9.43 8.95 (2.7) 10.63 21.32
2nd trimester 1,934 2.14 7.41 9.56 8.99 (2.74) 10.68 18.65
3rd trimester 1,920 2.26 7.41 9.51 9.09 (2.93) 10.97 20.33
Overall pregnancy 1,932 2.14 7.69 9.56 9 (2.32) 10.84 13.77
0–2 y 1,894 3.03 7.41 9.54 8.75 (2.01) 10.28 12.04
2–4 y 1,763 2.61 7.15 9.14 8.38 (1.79) 9.62 11.61
Note: CANDLE, Conditions Affecting Neurocognitive Development and Learning in Early Childhood; ECHO, Environmental Influences on Child Health Outcomes; GAPPS, Global Alliance to Prevent Prematurity and Stillbirth; min, minimum; max, maximum; SD, standard deviation; TIDES, The Infant Development and Environment Study.

Associations of Air Pollution Exposures with Child Total Problems Score and IQ

N O 2 .

Higher exposures to NO 2 in the first trimester [ β : 0.70; (95% confidence interval (CI): 0.13, 1.27) per 2  ppb  NO 2 ], the second trimester [ β : 0.92 (95% CI: 0.31, 1.53) per 2  ppb  NO 2 ], and averaged over the whole pregnancy [ β : 1.24 (95% CI: 0.39, 2.08) per 2  ppb  NO 2 ] were associated with more behavioral problems in children (Table 4). There was no significant association between prenatal NO 2 and child IQ, and we found no significant association between postnatal NO 2 and either outcome.
Table 4 Associations of NO 2 and PM 2.5 in each pre- and postnatal window with child total problems score and IQ estimated from multivariable linear regressions in the overall analytic sample from the three U.S. pregnancy cohorts in the ECHO-PATHWAYS consortium (the CANDLE, TIDES, and GAPPS study).
Modelb NO 2 a PM 2.5 a
Total problems score IQ Total problems score IQ
n c β (95% CI) n c β (95% CI) n c β (95% CI) n c β (95% CI)
1st trimester
 Model 1 1,823 0.8 (0.32, 1.28) 1,776 0.8 ( 1.17 , 0.42 ) 1,823 1.07 (0.1, 2.04) 1,776 0.67 ( 1.45 , 0.11)
 Model 2 1,376 0.7 (0.13, 1.27) 1,423 0.28 ( 0.1 , 0.66) 1,376 1.32 (0.12, 2.52) 1,423 0.8 ( 0.01 , 1.62)
 Model 3 1,347 0.58 ( 0.02 , 1.17) 1,391 0.37 ( 0.03 , 0.77) 1,347 1.28 (0.08, 2.48) 1,391 0.89 (0.05, 1.73)
2nd trimester
 Model 1 1,822 0.94 (0.4, 1.48) 1,775 0.95 ( 1.33 , 0.56 ) 1,822 0.58 ( 0.36 , 1.52) 1,775 1.22 ( 1.9 , 0.53 )
 Model 2 1,376 0.92 (0.31, 1.53) 1,423 0.15 ( 0.24 , 0.54) 1,376 0.55 ( 0.6 , 1.71) 1,423 0.62 ( 1.36 , 0.12)
 Model 3 1,347 0.94 (0.3, 1.59) 1,391 0.16 ( 0.25 , 0.57) 1,347 0.41 ( 0.83 , 1.65) 1,391 0.48 ( 1.27 , 0.31)
3rd trimester
 Model 1 1,811 0.34 ( 0.15 , 0.82) 1,764 1.31 ( 1.71 , 0.91 ) 1,811 0.25 ( 0.64 , 1.15) 1,764 1.76 ( 2.42 , 1.1 )
 Model 2 1,368 0.27 ( 0.31 , 0.84) 1,415 0.25 ( 0.64 , 0.14) 1,368 0.54 ( 1.48 , 0.41) 1,415 0.33 ( 0.98 , 0.32)
 Model 3 1,339 0.3 ( 0.29 , 0.9) 1,383 0.27 ( 0.67 , 0.13) 1,339 0.99 ( 2.05 , 0.08) 1,383 0.17 ( 0.88 , 0.54)
Overall pregnancy
 Model 1 1,821 1.22 (0.54, 1.9) 1,774 1.59 ( 2.08 , 1.1 ) 1,821 1.81 (0.25, 3.37) 1,774 3.52 ( 4.72 , 2.32 )
 Model 2 1,376 1.24 (0.39, 2.08) 1,423 0.13 ( 0.37 , 0.63) 1,376 1.38 ( 0.6 , 3.35) 1,423 0.26 ( 1.53 , 1.01)
 Model 3 1,347 1.22 (0.34, 2.09) 1,391 0.17 ( 0.35 , 0.7) 1,347 1.03 ( 1.27 , 3.34) 1,391 0.18 ( 1.25 , 1.62)
0–2 y
 Model 1 1,792 0.7 ( 0.03 , 1.43) 1,741 1.55 ( 2.14 , 0.96 ) 1,792 2.09 (0.01, 4.16) 1,741 6.03 ( 7.8 , 4.25 )
 Model 2 1,363 0.41 ( 0.53 , 1.34) 1,407 0.37 ( 0.21 , 0.95) 1,363 2.55 ( 0.16 , 5.27) 1,407 1.47 ( 3.4 , 0.46)
 Model 3 1,334 0.67 ( 0.28 , 1.62) 1,375 0.25 ( 0.35 , 0.85) 1,334 1.62 ( 1.29 , 4.53) 1,375 0.8 ( 3 , 1.41)
2–4 y
 Model 1 1,783 0.63 ( 0.07 , 1.34) 1,741 1.51 ( 2.07 , 0.94 ) 1,691 3.45 (1.24, 5.67) 1,622 8.31 ( 10.33 , 6.29 )
 Model 2 1,347 0.32 ( 0.57 , 1.21) 1,393 0.06 ( 0.49 , 0.61) 1,287 3.59 (0.35, 6.84) 1,311 2.63 ( 5.08 , 0.17 )
 Model 3 1,318 0.44 ( 0.46 , 1.34) 1,361 0 ( 0.56 , 0.57) 1,262 2.55 ( 0.82 , 5.92) 1,284 2.18 ( 5 , 0.64)
Note: BMI, body mass index; CANDLE, Conditions Affecting Neurocognitive Development and Learning in Early Childhood; CBCL, Child Behavior Checklist; CI, confidence interval; ECHO, Environmental Influences on Child Health Outcomes; GAPPS, Global Alliance to Prevent Prematurity and Stillbirth; IQ, intelligence quotient; TIDES, The Infant Development and Environment Study.
a
NO 2 and PM 2.5 in each window were rescaled to 2-unit increments.
b
Multivariable linear regressions were performed. Model 1 (the minimal model) minimally controlled for child sex, child age at outcome assessments, and study site. An indicator of CBCL forms was additionally included in the analysis of Total Problems score. Model 2 (the primary model) was further adjusted for child race, maternal education, log-transformed region- and inflation-adjusted household income, household members, an interaction between household members and income, marital status, maternal age at delivery, birth order, pregnancy smoking, pregnancy alcohol consumption, maternal depression, maternal IQ, child secondhand smoking exposure, and Child Opportunity Index (the domains of educational and economic opportunity) in corresponding windows with PM 2.5 and NO 2 exposures. Model 3 (the extended model) included additional adjustments for prepregnancy BMI, pregnancy supplement intakes, breastfeeding, and child year of birth.
c
n is the analytic sample size for each model.

P M 2.5 .

We found an adverse association between PM 2.5 in the first trimester and behavioral functioning [ β : 1.32 (95% CI: 0.12, 2.52)], but an insignificantly positive association with child IQ [ β : 0.80 (95% CI: 0.01 , 1.62)]. For postnatal exposures, each 2 - μ g / m 3 increase in PM 2.5 at age 2–4 y was associated with a 3.59 unit (95% CI: 0.35, 6.84) higher Total Problems score and a 2.63 point (95% CI: 5.08 , 0.17 ) lower child IQ. Additionally, children with higher PM 2.5 exposures at age 0–2 y were estimated to have a 2.55-unit (95% CI: 0.16 , 5.27) higher Total Problems score, and a 1.47-point (95% CI: 3.40 , 0.46) lower IQ on average, in comparison with their counterparts with a 2 - μ g / m 3 lower exposure; however, these results had larger statistical uncertainty.

Sex Modification

From the interaction models with child sex, we found a stratum-specific association between higher second trimester PM 2.5 and more behavioral problems in girls [ β : 1.50 (95% CI: 0.19, 2.81) in girls, β : 0.35 (95% CI: 1.89 , 1.18) in boys, P interaction : 0.026], and a stratum-specific association between higher second trimester PM 2.5 and a lower IQ in boys [ β : 0.07 (95% CI: 0.92 , 0.79) in girls, β : 1.19 (95% CI: 2.18 , 0.2 ) in boys, P interaction : 0.040], although the results were null from the primary analysis (Figure 2; Table S3). In both postnatal windows, there was suggestive evidence of stronger estimated effects of PM 2.5 on Total Problems score in girls than in boys; nevertheless, the insignificant interaction terms indicated that the effect difference between groups might be due to chance [0–2 y: β : 3.50 (95% CI: 0.61, 6.39) in girls, β : 1.72 (95% CI: 1.25 , 4.70) in boys, P interaction : 0.101; 2–4 y: β : 4.80 (95% CI: 1.25, 8.36) in girls, β : 2.74 (95% CI: 0.74 , 6.23) in boys, P interaction : 0.121]. No sex difference in other associations was detected.
Figure 2. Shown are the estimated effects of air pollution exposures on child Total Problems score and IQ by child sex (male vs. female) in participants from the three U.S. pregnancy cohorts in the ECHO-PATHWAYS Consortium (the CANDLE, TIDES, and GAPPS study). NO 2 and PM 2.5 in each window were rescaled to 2-unit increments. In addition to the interaction term between individual air pollution exposures in each window and child sex, the linear regressions were adjusted for child sex, child age at outcome assessments, study site, child race, maternal education, log-transformed region- and inflation-adjusted household income, household members, an interaction between household members and income, marital status, maternal age at delivery, birth order, pregnancy smoking, pregnancy alcohol consumption, maternal depression, maternal IQ, child secondhand smoking exposure, and Child Opportunity Index (the domains of educational and economic opportunity) in corresponding windows with PM 2.5 and NO 2 exposures. An indicator of CBCL forms was additionally included in the analysis of Total Problems score. The p -value indicates the statistical significance of the interaction term. The symbols of triangles and circles indicate the effect estimate, the error bars show 95% confidence intervals, and the dotted lines show null values. Numeric data (including sample size for each association) are shown in Table S3. Note: CANDLE, Conditions Affecting Neurocognitive Development and Learning in Early Childhood; CBCL, Child Behavior Checklist; ECHO, Environmental Influences on Child Health Outcomes; GAPPS, Global Alliance to Prevent Prematurity and Stillbirth; IQ, intelligence quotient; TIDES, The Infant Development and Environment Study.

Sensitivity Analyses

First, to handle the missing data in all covariates in the primary model, we constructed the ROC curve to verify the plausibility of the MAR assumption. An average area under curve of 0.891 (95% CI: 0.868, 0.914) was calculated, suggesting that we had an 89.1% chance of correctly distinguishing a complete case from one with missing data using the model with only covariates (Figure S4). We then implemented MICE to impute the missingness and show the results in Table S4. In comparison with the primary results, the point estimates obtained from MICE for the associations with child Total Problems score were mostly attenuated with a higher precision, whereas there was no clear pattern for the changes in associations with child IQ. The positive associations between NO 2 in the first trimester, the second trimester, and the whole pregnancy and Total Problems score achieved statistical significance in both the complete case analysis and MICE [first trimester: β : 0.51 (95% CI: 0.03, 0.98); second trimester: β : 0.61 (95% CI: 0.11, 1.11); whole pregnancy: β : 0.81 (95% CI: 0.17, 1.45)]. The positive association between first trimester PM 2.5 and IQ also gained a higher precision in MICE [ β : 0.70 (95% CI: 0.05, 1.35)]. Second, we estimated a correlation of 0.99 between the raw Total Problems score and the standardized z-score, and the conclusions remained the same after we replaced the standardized z-score as the outcome (Table S5). Third, when restricting the analytic sample to participants born at gestational week 34 or later, the estimated direct effects of air pollution exposures on both outcomes were very similar to the findings from the primary analysis except for a slightly larger association with significance between first trimester PM 2.5 and IQ [ β : 0.88 (95% CI: 0.06, 1.70)] (Table S6). Fourth, after simultaneously adjusting for exposures across windows, we derived significant associations between second trimester NO 2 and Total Problems score with smaller effect sizes than those from single exposure models [adjustments for NO 2 in other trimesters: β : 0.77 (95% CI: 0.01, 1.53); adjustments for NO 2 in other trimesters and postnatal windows: β : 0.79 (95% CI: 0.04, 1.54)] (Table S7). Stronger positive associations between first trimester PM 2.5 and IQ were also detected when PM 2.5 in other windows were included [adjustments for PM 2.5 in other trimesters: β : 0.84 (95% CI: 0.02, 1.66); adjustments for PM 2.5 in other trimesters and postnatal windows: β : 1.29 (95% CI: 0.30, 2.29)]. Other significant associations in the primary analysis were attenuated to null. Constrained DLM results depended on the df and were generally not consistent with our primary findings. The DLM with a df of 8 identified a critical window of gestational week 4–5 where higher PM 2.5 exposures were related to more behavioral problems (Figure S5), which agreed with the significantly positive association found in the first trimester. DLMs using a df of 4 to 9 also indicated positive associations between PM 2.5 and IQ in slightly different windows within the range of gestation week 6–13, and a similar relationship has been evident in the analysis with the exposure in the first trimester, although the results are insignificant. Apart from these findings, DLMs either showed associations that contradicted our hypotheses and the primary results or displayed associations that were null in our primary analysis, particularly in DLMs with a high df. In addition, the smooth effect curves generated from GAMs in the overall sample (Figure 3) and in each sex stratum (Figure S6) generally did not indicate significant departures from the conclusions of the primary and secondary analyses, although the association between first trimester PM 2.5 and Total Problems score appeared to be predominantly driven by the exposure outliers in the high end. Last, leaving out the CANDLE cohort (Memphis) presented the biggest impact on results of the leave-one-cohort-out (Table S8) and leave-one-site-out (Table S9) analyses: All the significant associations detected in the primary analysis became null. However, the exclusion of most other study sites did not cause meaningful changes in the result. Comparing the fully adjusted fixed effects models (i.e., the primary analysis) to fixed effects models with site–covariate interactions as well as mixed-effects models both with and without site–covariate interactions, results were generally consistent. We found attenuation in the association of first trimester PM 2.5 with Total Problems score and the associations of PM 2.5 in age 2–4 y with both outcomes in the mixed-effects models, but inclusion of site–covariate interactions corrected these attenuations, raising the possibility of site-specific confounding that is only apparent when one of the sites with a smaller population is upweighted in the mixed-effects model (Table S10).
Figure 3. Shown are the graphic examination of the linearity of associations of NO 2 and PM 2.5 in each window with child Total Problems score and IQ from the fully adjusted generalized additive models in the overall analytic sample of three cohorts. The models were controlled for child sex, child age at outcome assessments, study site, child race, maternal education, log-transformed region- and inflation-adjusted household income, household members, an interaction between household members and income, marital status, maternal age at delivery, birth order, pregnancy smoking, pregnancy alcohol consumption, maternal depression, maternal IQ, child secondhand smoking exposure, and Child Opportunity Index (the domains of educational and economic opportunity) in corresponding windows with PM 2.5 and NO 2 exposures. An indicator of CBCL forms was additionally included in the analysis of Total Problems score. The p -value indicates the statistical significance of the association. The n indicates the analytic sample size. Black solid lines represent the potential nonlinear associations, gray bands are 95% CIs, and red dotted lines show null values. Note: CANDLE, Conditions Affecting Neurocognitive Development and Learning in Early Childhood; CBCL, Child Behavior Checklist; CI, confidence interval; ECHO, Environmental Influences on Child Health Outcomes; GAPPS, Global Alliance to Prevent Prematurity and Stillbirth; IQ, intelligence quotient; TIDES, The Infant Development and Environment Study.

Discussion

We used a large, combined sample from three sociodemographically diverse pregnancy cohorts situated in six U.S. cities to examine the associations of the two regulated air pollutants— NO 2 and PM 2.5 —with child behavioral problems and cognitive performance at age 4–6 y. Children whose mothers experienced higher NO 2 exposures during pregnancy, particularly in the first and second trimester, were more likely to have behavioral problems. Associations between prenatal NO 2 and child IQ or postnatal NO 2 with either outcome were not evident. We also found a positive association of first trimester PM 2.5 with Total Problems score; nevertheless, this association needs to be interpreted with caution because it may be driven by outliers. In addition, higher exposures to postnatal PM 2.5 when children were 2–4 y were associated with poorer child behavioral functioning and cognitive performance. The associations between PM 2.5 and Total Problems score were generally more pronounced in girls, and the inverse association between second trimester PM 2.5 and IQ was detected only in boys. Conclusions remained largely unchanged with expanded covariate adjustments and in most sensitivity analyses, but DLMs failed to confirm most critical windows being identified.
The adverse associations between prenatal NO 2 exposure and child behavior, particularly our findings for exposures in early- to midpregnancy, align with similar evidence from several previous studies. A previous CANDLE cohort study reported a 6% increased risk of externalizing behavior for each 2 -ppb higher prenatal NO 2 .39 A study in Japan found increased odds of attention problems and aggressive behaviors in children with higher prenatal NO 2 exposures.34 Similarly, another study by Ren et al. (2019) reported a positive association between prenatal exposures to NO 2 and total difficulties and suggested that NO 2 in the first trimester may be more deleterious.32 Moreover, there is increasing evidence from population studies linking prenatal air pollution exposures, including NO 2 , with attention deficit hyperactivity disorder (ADHD) and autism spectrum disorders.10,11 Prenatal air pollution may interfere with fetal neurodevelopment by inducing oxidative stress and inflammation or altering the epigenetic programming in the placenta or fetus.1214,81,82 However, we observed null associations between postnatal NO 2 and child behaviors, which was inconsistent with our previous finding in CANDLE,39 as well as the two studies combining multiple cohorts in Spain.30,83 Our results for the association between NO 2 exposures in either prenatal or postnatal windows and child IQ were also null, paralleling those from the prior CANDLE study using NO 2 estimates from a national annual model,27 two other studies in Europe,43,84 and one study in Taiwan.36 However, significant adverse associations were reported in five other European studies.31,41,8587 The disparity in findings may be driven by variations in the air pollution prediction models, the exposure levels, the exposure duration (short-term vs. long-term), the exposure locations (school vs. home), the source of exposure (indoor vs. outdoor), outcome assessment, underlying susceptibility in study populations, or confounder selection.
The detected positive relationship of first trimester PM 2.5 with child Total Problems score was in agreement with findings from several existing studies with prenatal PM exposures in mainland China,32 Japan,34 Korea,33 and Mexico City.37 Some of our sensitivity analyses suggested a potential positive association between first trimester PM 2.5 and IQ, such as MICE and DLM, although the result was insignificant in the primary analysis. We only know of two previous studies measuring the effect of PM in specific trimester(s) on child cognitive performance. A study in Massachusetts detected no association between third trimester PM 2.5 and child IQ,28 though the associations with first and second trimester exposures were not evaluated, and a study in Taiwan reported a null relationship of first trimester PM 10 and child neurodevelopmental scores at 6 and 18 months.36 Using a distributed lag modeling approach, another study showed that children in Boston with higher PM 2.5 exposure at 31–38 gestational weeks had a lower IQ at age 6,25 the critical windows of which differed from our findings from DLM. This positive association between first trimester PM 2.5 and IQ contrasts with our hypothesis, and its interpretation may reflect the following considerations: first, this protective association may suggest potential selection bias from multiple sources. One is the enrollment criterion of women with low medical risk pregnancies in the CANDLE and TIDES cohort, and another is the fact that the outcomes are conditioned on live birth.88,89 When the analytic sample was restricted to participants with a gestational week of 34 and above, this association became stronger, indicating that constraint on gestational age may also induce bias. Second, this result was largely attenuated when we excluded participants in Yakima, Washington, from the analysis. A potential explanation is that the spatiotemporal model may generate less accurate predictions in such a region where wildfires and agricultural burning are major sources of particulate pollution.
The associations with the greatest magnitude in our analysis were found between postnatal PM 2.5 and child behavior, particularly exposures at age 2–4 y. A similar conclusion was drawn in a German study by Fuertes et al. (2016),29 which reported an increased risk of hyperactivity/inattention in adolescents with higher exposures of PM 2.5 mass and absorbance at 10 y and 15 y address. However, in an analysis from the Project Viva cohort, significant associations between postnatal PM 2.5 averaged in different windows and teacher-rated behavioral problems and executive functions were detected only in minimally adjusted models.90 Our estimated effects of postnatal PM 2.5 on child IQ were also relatively strong. This result is consistent with two studies based on multiple cohorts in Spain, which reported a reduced growth in work memory among children 7–11 y of age with higher PM 2.5 exposure from commutes or at school.30,83 Nevertheless, a study in Upstate New York with relatively low air pollution exposures showed mixed associations between PM 2.5 assessments and risk of failure on developmental screening using Ages and Stages Questionnaires at 8 to 36 months of age.26 Another study in Massachusetts found a null association between PM 2.5 in early childhood and child IQ at age 8 y.28 In comparison with younger children, 2- to 4-y-old children are more likely to stay outdoor for longer periods and are usually more active, which may increase their susceptibility to air pollution.91 According to a conceptual framework proposed by Tulve et al. (2016), postnatal air pollution interplays with inherent characteristics, activities and behaviors, and other stressors from built, natural, and social environments and influences child learning, communication, response to stress, and general psychological well-being.92 In addition, laboratory and imaging studies have shown that the number of neural connections explodes in the first and second year of life,93 and brain size increases 4-fold, reaching 90% of adult volume by age 6 y.1,9497 Inhaled particles can invade deep in the lung and translocate along the olfactory nerve into the olfactory bulb.22 A pilot study in healthy children and young dogs similarly exposed to high air pollution in Mexico City showed a significant up-regulation of inflammatory markers and histological changes in target brain areas.98 Studies in animals also observed inflammatory responses in the prefrontal cortex and the striatum after air pollution exposure—the regions related to executive functions such as working memory.99103 These findings provide strong mechanistic support for the hypothesis that inhaled air pollutants may trigger oxidative stress and promote inflammatory markers across the impaired blood brain barrier, which result in microglial activation and elevated cytokine expression, and in turn cause CNS damage relevant for behavioral and cognitive function.
Our results somewhat suggested stronger associations between PM 2.5 and child behavioral functioning in girls, particularly with postnatal PM 2.5 . Neither the study in Germany29 nor the previous analysis in the CANDLE cohort39 found sex differences in the associations between postnatal PM and child behaviors. Nevertheless, the study in Mexico City reported a stronger association between first trimester PM 2.5 and reduced adaptive skills in boys. In addition, we found an inverse association between second trimester PM 2.5 and IQ only in boys, which agrees with much existing literature showing more pronounced findings in boys,25,30,35,4143 but disagrees with the three U.S. studies with null findings.26,27,104 Research has identified several sex differences in neurodevelopment, including morphological, physiological, and chemical differences.40 Although animal studies have shown that males are more susceptible to airborne metals than females are, which is potentially explained by sex-specific altered dopamine function,105 other evidence from laboratory science support a protective mechanism for boys via neuroprotective effects of androgens against oxidative stress.106
Our study has several important strengths. First, we combined three pregnancy cohorts into a large analytic sample with high sociodemographic diversity and controlled for several important confounders harmonized across cohorts, including individual and neighborhood SES indicators,107 maternal depression, and maternal IQ. The approach of pooling data helps leverage the spatiotemporal contrast in air pollution assessments and strengthens the external generalizability of the study results by increasing the diversity of participants. Second, we used spatiotemporally resolved air pollution predictions from a well-validated modeling approach based on individually geocoded residential addresses in six U.S. cities across multiple years, allowing us to exploit small-scale spatial variability in the pollutant surfaces over several windows in both pre- and postnatal periods. Last, we provided rigorous training for examiners and implemented robust protocols to collect standardized objective assessments of child cognitive performance. The data for both outcome measures were collected using standardized and validated neuropsychological testing tools and went through strict quality control.
There are also limitations to be acknowledged. One is the parent-report method for ascertaining child behaviors. Previous research has shown that parents report child psychological problems more often and of greater severity than teachers, suggesting combined reports from multiple sources may improve reliability.108 However, the study with participants from the Adolescent Brain Cognitive Development cohort in the United States found little psychometric evidence for maternal psychopathology biasing reports of child behavior problems.109 Use of parent report alone is common in epidemiological studies, given the ease of data collection. Another limitation is the heterogeneity among the three studies and sites in terms of exposure levels, air pollution compositions, frequency of address data collection, outcome assessment instruments, examiners, and measurement methods for covariates, which could induce measurement errors of various magnitudes. We performed several sensitivity analyses to investigate the impacts of certain heterogeneities on the detected associations in a pooled sample. Although the results from our leave-one-cohort-out and leave-one-site-out analyses indicated that the CANDLE study contributed the most to the findings, likely due to its large sample size, the comparisons between fixed-effects models and mixed-effects models with or without site–covariate interactions suggested that the roles of site heterogeneity and potential site-specific confounding on the estimated associations were relatively minor. The third concern is the potential inaccuracy in air pollution assessments. Our prediction model, like other modeling approaches, may produce complex forms of measurement error that can distort the true associations.110,111 The current analysis did not account for indoor exposures or exposures in the other locations, such as day care, preschool, or daily commutes. We also lacked other air pollutants that were linked with child neurodevelopment in previous research, such as black carbon, ozone, PAHs, and sulfur dioxide.10,11 Moreover, collinearity of exposures across windows may cause inaccurate identification of critical window; we thus implemented constrained DLM to verify our results. However, the results from DLMs were largely unmatched with our primary findings. Defining exposure windows a priori is particularly appealing, because the results are easy to interpret and to compare, and the evidence can be used to inform interventions directly. Certain clinical problems are likely to cluster in different trimesters, such as teratogenesis112 or miscarriage113 in the first trimester and bleeding114 in the third trimester. We also expect tremendous physical, behavioral, social, and emotional advancements to occur in children when they turn age 2. However, these predefined windows may not reflect many important developmental milestones115 nor correspond to relevant vulnerable periods of neurodevelopmental impairments. On the contrary, the results from DLMs are very sensitive to model specification, and they could generate spurious significant windows or fail to capture windows when the smoothness is imposed incorrectly.116 In such manner, neither method has generated completely valid conclusions, and the sensitive exposure periods identified by our analysis merit further study. Furthermore, we were missing 16% of the maternal IQ measurements. Based on our assessment of the ROC curve, the assumption of missing at random was likely valid, and the results from both the complete cases analysis and the multiple imputation were considered robust.75,117 Because multiple imputation may not be readily compatible with DLMs or generalized additive models, we employed it as an alternative approach to verify the findings from the complete cases analysis. Nevertheless, we cannot rule out potential selection bias with confidence, given the discrepancy of results from the two analytic approaches. In addition, residual confounding may exist. Previous studies found adverse individual and joint neurobehavioral associations with transportation noise and traffic-related air pollution in children,31,118 but we did not control for noise due to data unavailability. Last, our findings may need to be interpreted with caution owing to the multiple comparisons.
Despite these limitations, our study extends earlier findings that have raised concern of reduced behavioral functioning and cognitive performance in children following NO 2 and PM 2.5 exposures in early life. We used highly refined exposure assessments across several pre- and postnatal windows in U.S. settings with modest air pollution levels. Aside from filling the methodological gaps in the current literature, our study explores the most relevant exposure window, compares the findings across two neurodevelopmental measures, and highlights the extra vulnerability to different neurodevelopmental impairments in each sex when exposed to air pollution. Enhanced understanding of population vulnerabilities to common ambient air pollutants are necessary to ensure that regulatory policies provide adequate protection for all.

Acknowledgments

This research is supported by the ECHO-PATHWAYS consortium [National Institutes of Health (NIH): 1UG3OD023271, 4UH3OD023271, 1R01HL109977, and P30ES007033]. The CANDLE study is funded by both the Urban Child Institute and NIH (1R01HL109977). The TIDES study is funded by NIH (TIDES I: R01ES01686; TIDES II: 1R01ES25169). Air pollution models were developed under STAR research assistance agreements, No. RD831697 (MESA Air) and RD-83830001 (MESA Air Next Stage), RD83479601 (University of Washington Center for Clean Air Research), and R83374101 (MESA Coarse), awarded by the U.S. Environmental Protection Agency (EPA), with additional grants R56ES026528, R01ES023500, and R01ES025888 from the National Institute of Environmental Health Sciences (NIEHS) and P01AG055367 from the National Institute on Aging (NIA). Funding was also provided by Kresge Foundation Grant No. 243365. E.B. is supported by the NIH (P30 ES005022). M.H. is supported in part by the University of Washington NIEHS sponsored Biostatistics, Epidemiologic and Bioinformatic Training in Environmental Health (BEBTEH) Training Grant (NIEHS T32ES015459). This manuscript has not been formally reviewed by the U.S. EPA. The views expressed in this document are solely those of the authors, and the U.S. EPA does not endorse any products or commercial services mentioned in this publication.
The data used for this study are not publicly available, but deidentified data may be available on request, subject to approval by the internal review board and under a formal data use agreement. Contact the corresponding author for more information. The computing code in R can be obtained from the corresponding author via email request.

Article Notes

A.A.S. has been a consultant for the Health Effects Institute on the traffic-related air pollution panel. E.B. was a consultant for the California Environmental Protection Agency in 2020. Their freedom to design, conduct, interpret and publish research is not compromised by the institutes. The other authors declare they have no actual or potential competing financial interests.

Supplementary Material

File (ehp10248.smcontents.508.pdf)
File (ehp10248.s001.acco.pdf)

References

1.
Stiles J, Jernigan TL. 2010. The basics of brain development. Neuropsychol Rev 20(4):327–348. https://pubmed.ncbi.nlm.nih.gov/21042938/, https://doi.org/10.1007/s11065-010-9148-4.
2.
Stiles J. 2008. The Fundamentals of Brain Development: Integrating Nature and Nurture. 1st edition. Harvard University Press.
3.
Bystron I, Blakemore C, Rakic P. 2008. Development of the human cerebral cortex: Boulder Committee revisited. Nat Rev Neurosci 9(2):110–122. https://pubmed.ncbi.nlm.nih.gov/18209730/, https://doi.org/10.1038/nrn2252.
4.
Lenroot RK, Giedd JN. 2006. Brain development in children and adolescents: insights from anatomical magnetic resonance imaging. Neurosci Biobehav Rev 30(6):718–729. https://pubmed.ncbi.nlm.nih.gov/16887188/, https://doi.org/10.1016/j.neubiorev.2006.06.001.
5.
Iwasaki N, Hamano K, Okada Y, Horigome Y, Nakayama J, Takeya T, et al. 1997. Volumetric quantification of brain development using MRI. Neuroradiology 39(12):841–846. https://pubmed.ncbi.nlm.nih.gov/9457706/, https://doi.org/10.1007/s002340050517.
6.
Hofstra MB, Van der Ende J, Verhulst FC. 2000. Continuity and change of psychopathology from childhood into adulthood: a 14-year follow-up study. J Am Acad Child Adolesc Psychiatry 39(7):850–858. https://pubmed.ncbi.nlm.nih.gov/10892226/, https://doi.org/10.1097/00004583-200007000-00013.
7.
Shaw DS, Gilliom M, Ingoldsby EM, Nagin DS. 2003. Trajectories leading to school-age conduct problems. Dev Psychol 39(2):189–200. https://pubmed.ncbi.nlm.nih.gov/12661881/, https://doi.org/10.1037//0012-1649.39.2.189.
8.
Althoff RR, Verhulst FC, Rettew DC, Hudziak JJ, van der Ende J. 2010. Adult outcomes of childhood dysregulation: a 14-year follow-up study. J Am Acad Child Adolesc Psychiatry 49(11):1105–1116. https://pubmed.ncbi.nlm.nih.gov/20970698/, https://doi.org/10.1016/j.jaac.2010.08.006.
9.
Clifford A, Lang L, Chen R, Anstey KJ, Seaton A. 2016. Exposure to air pollution and cognitive functioning across the life course–a systematic literature review. Environ Res 147:383–398. https://pubmed.ncbi.nlm.nih.gov/26945620/, https://doi.org/10.1016/j.envres.2016.01.018.
10.
Volk HE, Perera F, Braun JM, Kingsley SL, Gray K, Buckley J, et al. 2021. Prenatal air pollution exposure and neurodevelopment: a review and blueprint for a harmonized approach within ECHO. Environ Res 196:110320. https://pubmed.ncbi.nlm.nih.gov/33098817/, https://doi.org/10.1016/j.envres.2020.110320.
11.
Suades-González E, Gascon M, Guxens M, Sunyer J. 2015. Air pollution and neuropsychological development: a review of the latest evidence. Endocrinology 156(10):3473–3482. https://pubmed.ncbi.nlm.nih.gov/26241071/, https://doi.org/10.1210/en.2015-1403.
12.
Nachman RM, Mao G, Zhang X, Hong X, Chen Z, Soria CS, et al. 2016. Intrauterine inflammation and maternal exposure to ambient PM2.5 during preconception and specific periods of pregnancy: the Boston Birth Cohort. Environ Health Perspect 124(10):1608–1615. https://pubmed.ncbi.nlm.nih.gov/27120296/, https://doi.org/10.1289/EHP243.
13.
Hogervorst JGF, Madhloum N, Saenen ND, Janssen BG, Penders J, Vanpoucke C, et al. 2019. Prenatal particulate air pollution exposure and cord blood homocysteine in newborns: results from the ENVIRONAGE birth cohort. Environ Res 168:507–513. https://pubmed.ncbi.nlm.nih.gov/30477822/, https://doi.org/10.1016/j.envres.2018.08.032.
14.
Grandjean P, Landrigan PJ. 2006. Developmental neurotoxicity of industrial chemicals. Lancet 368(9553):2167–2178. https://pubmed.ncbi.nlm.nih.gov/17174709/, https://doi.org/10.1016/S0140-6736(06)69665-7.
15.
Bové H, Bongaerts E, Slenders E, Bijnens EM, Saenen ND, Gyselaers W, et al. 2019. Ambient black carbon particles reach the fetal side of human placenta. Nat Commun 10(1):3866. https://pubmed.ncbi.nlm.nih.gov/31530803/, https://doi.org/10.1038/s41467-019-11654-3.
16.
Campbell A. 2004. Inflammation, neurodegenerative diseases, and environmental exposures. Ann NY Acad Sci 1035:117–132. https://pubmed.ncbi.nlm.nih.gov/15681804/, https://doi.org/10.1196/annals.1332.008.
17.
Saenen ND, Plusquin M, Bijnens E, Janssen BG, Gyselaers W, Cox B, 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(8):834–840. https://pubmed.ncbi.nlm.nih.gov/25816123/, https://doi.org/10.1289/ehp.1408549.
18.
Calderón-Garcidueñas L, Calderón-Garcidueñas A, Torres-Jardón R, Avila-Ramírez J, Kulesza RJ, Angiulli AD. 2015. Air pollution and your brain: what do you need to know right now. Prim Health Care Res Dev 16(04):329–345. https://pubmed.ncbi.nlm.nih.gov/25256239/, https://doi.org/10.1017/S146342361400036X.
19.
Davis DA, Bortolato M, Godar SC, Sander TK, Iwata N, Pakbin P, et al. 2013. Prenatal exposure to urban air nanoparticles in mice causes altered neuronal differentiation and depression-like responses. PLoS One 8(5):e64128. https://pubmed.ncbi.nlm.nih.gov/23734187/, https://doi.org/10.1371/journal.pone.0064128.
20.
Frye RE, Cakir J, Rose S, Delhey L, Bennuri SC, Tippett M, et al. 2021. Prenatal air pollution influences neurodevelopment and behavior in autism spectrum disorder by modulating mitochondrial physiology. Mol Psychiatry 26(5):1561–1577. https://pubmed.ncbi.nlm.nih.gov/32963337/, https://doi.org/10.1038/s41380-020-00885-2.
21.
Peters A, Veronesi B, Calderón-Garcidueñas L, Gehr P, Chen LC, Geiser M, et al. 2006. Translocation and potential neurological effects of fine and ultrafine particles a critical update. Part Fibre Toxicol 3:13. https://pubmed.ncbi.nlm.nih.gov/16961926/, https://doi.org/10.1186/1743-8977-3-13.
22.
Oberdörster G, Sharp Z, Atudorei V, Elder A, Gelein R, Kreyling W, et al. 2004. Translocation of inhaled ultrafine particles to the brain. Inhal Toxicol 16(6–7):437–445. https://pubmed.ncbi.nlm.nih.gov/15204759/, https://doi.org/10.1080/08958370490439597.
23.
Block ML, Wu X, Pei Z, Li G, Wang T, Qin L, et al. 2004. Nanometer size diesel exhaust particles are selectively toxic to dopaminergic neurons: the role of microglia, phagocytosis, and NADPH oxidase. FASEB J 18(13):1618–1620. https://pubmed.ncbi.nlm.nih.gov/15319363/, https://doi.org/10.1096/fj.04-1945fje.
24.
Chen L, Yokel RA, Hennig B, Toborek M. 2008. Manufactured aluminum oxide nanoparticles decrease expression of tight junction proteins in brain vasculature. J Neuroimmune Pharmacol 3(4):286–295. https://pubmed.ncbi.nlm.nih.gov/18830698/, https://doi.org/10.1007/s11481-008-9131-5.
25.
Chiu Y-HM, Hsu H-HL, Coull BA, Bellinger DC, Kloog I, Schwartz J, et al. 2016. Prenatal particulate air pollution and neurodevelopment in urban children: examining sensitive windows and sex-specific associations. Environ Int 87:56–65. https://pubmed.ncbi.nlm.nih.gov/26641520/, https://doi.org/10.1016/j.envint.2015.11.010.
26.
Ha S, Yeung E, Bell E, Insaf T, Ghassabian A, Bell G, et al. 2019. Prenatal and early life exposures to ambient air pollution and development. Environ Res 174:170–175. https://pubmed.ncbi.nlm.nih.gov/30979514/, https://doi.org/10.1016/j.envres.2019.03.064.
27.
Loftus CT, Hazlehurst MF, Szpiro AA, Ni Y, Tylavsky FA, Bush NR, et al. 2019. Prenatal air pollution and childhood IQ: preliminary evidence of effect modification by folate. Environ Res 176:108505. https://pubmed.ncbi.nlm.nih.gov/31229778/, https://doi.org/10.1016/j.envres.2019.05.036.
28.
Harris MH, Gold DR, Rifas-Shiman SL, Melly SJ, Zanobetti A, Coull BA, et al. 2015. Prenatal and childhood traffic-related pollution exposure and childhood cognition in the Project Viva cohort (Massachusetts, USA). Environ Health Perspect 123(10):1072–1078. https://pubmed.ncbi.nlm.nih.gov/25839914/, https://doi.org/10.1289/ehp.1408803.
29.
Fuertes E, Standl M, Forns J, Berdel D, Garcia-Aymerich J, Markevych I, et al. 2016. Traffic-related air pollution and hyperactivity/inattention, dyslexia and dyscalculia in adolescents of the German GINIplus and LISAplus birth cohorts. Environ Int 97:85–92. https://pubmed.ncbi.nlm.nih.gov/27835751/, https://doi.org/10.1016/j.envint.2016.10.017.
30.
Alvarez-Pedrerol M, Rivas I, López-Vicente M, Suades-González E, Donaire-Gonzalez D, Cirach M, et al. 2017. Impact of commuting exposure to traffic-related air pollution on cognitive development in children walking to school. Environ Pollut 231(pt 1):837–844. https://pubmed.ncbi.nlm.nih.gov/28866425/, https://doi.org/10.1016/j.envpol.2017.08.075.
31.
van Kempen E, Fischer P, Janssen N, Houthuijs D, van Kamp I, Stansfeld S, et al. 2012. Neurobehavioral effects of exposure to traffic-related air pollution and transportation noise in primary schoolchildren. Environ Res 115:18–25. https://pubmed.ncbi.nlm.nih.gov/22483436/, https://doi.org/10.1016/j.envres.2012.03.002.
32.
Ren Y, Yao X, Liu Y, Liu S, Li X, Huang Q, et al. 2019. Outdoor air pollution pregnancy exposures are associated with behavioral problems in China’s preschoolers. Environ Sci Pollut Res Int 26(3):2397–2408. https://pubmed.ncbi.nlm.nih.gov/30467751/, https://doi.org/10.1007/s11356-018-3715-2.
33.
Kim E, Park H, Hong Y-C, Ha M, Kim Y, Kim B-N, et al. 2014. Prenatal exposure to PM10 and NO2 and children’s neurodevelopment from birth to 24 months of age: Mothers and Children’s Environmental Health (MOCEH) study. Sci Total Environ 481:439–445. https://pubmed.ncbi.nlm.nih.gov/24631606/, https://doi.org/10.1016/j.scitotenv.2014.01.107.
34.
Yorifuji T, Kashima S, Diez MH, Kado Y, Sanada S, Doi H. 2017. Prenatal exposure to outdoor air pollution and child behavioral problems at school age in Japan. Environ Int 99:192–198. https://pubmed.ncbi.nlm.nih.gov/27890345/, https://doi.org/10.1016/j.envint.2016.11.016.
35.
Chiu Y-HM, Bellinger DC, Coull BA, Anderson S, Barber R, Wright RO, et al. 2013. Associations between traffic-related black carbon exposure and attention in a prospective birth cohort of urban children. Environ Health Perspect 121(7):859–864. https://pubmed.ncbi.nlm.nih.gov/23665743/, https://doi.org/10.1289/ehp.1205940.
36.
Lin C-C, Yang S-K, Lin K-C, Ho W-C, Hsieh W-S, Shu B-C, et al. 2014. Multilevel analysis of air pollution and early childhood neurobehavioral development. Int J Environ Res Public Health 11(7):6827–6841. https://pubmed.ncbi.nlm.nih.gov/24992486/, https://doi.org/10.3390/ijerph110706827.
37.
McGuinn LA, Bellinger DC, Colicino E, Coull BA, Just AC, Kloog I, et al. 2020. Prenatal PM2.5 exposure and behavioral development in children from Mexico City. Neurotoxicology 81:109–115. https://pubmed.ncbi.nlm.nih.gov/32950567/, https://doi.org/10.1016/j.neuro.2020.09.036.
38.
Sunyer J, Suades-González E, García-Esteban R, Rivas I, Pujol J, Alvarez-Pedrerol M, et al. 2017. Traffic-related air pollution and attention in primary school children: short-term association. Epidemiology 28(2):181–189. https://pubmed.ncbi.nlm.nih.gov/27922536/, https://doi.org/10.1097/EDE.0000000000000603.
39.
Loftus CT, Ni Y, Szpiro AA, Hazlehurst MF, Tylavsky FA, Bush NR, et al. 2020. Exposure to ambient air pollution and early childhood behavior: a longitudinal cohort study. Environ Res 183:109075. https://pubmed.ncbi.nlm.nih.gov/31999995/, https://doi.org/10.1016/j.envres.2019.109075.
40.
Andreano JM, Cahill L. 2009. Sex influences on the neurobiology of learning and memory. Learn Mem 16(4):248–266. https://pubmed.ncbi.nlm.nih.gov/19318467/, https://doi.org/10.1101/lm.918309.
41.
Sunyer J, Esnaola M, Alvarez-Pedrerol M, Forns J, Rivas I, López-Vicente M, et al. 2015. Association between traffic-related air pollution in schools and cognitive development in primary school children: a prospective cohort study. PLoS Med 12(3):e1001792. https://pubmed.ncbi.nlm.nih.gov/25734425/, https://doi.org/10.1371/journal.pmed.1001792.
42.
Cowell WJ, Bellinger DC, Coull BA, Gennings C, Wright RO, Wright RJ. 2015. Associations between prenatal exposure to black carbon and memory domains in urban children: modification by sex and prenatal stress. PLoS One 10(11):e0142492. https://pubmed.ncbi.nlm.nih.gov/26544967/, https://doi.org/10.1371/journal.pone.0142492.
43.
Lertxundi A, Andiarena A, Martínez MD, Ayerdi M, Murcia M, Estarlich M, et al. 2019. Prenatal exposure to PM2.5 and NO2 and sex-dependent infant cognitive and motor development. Environ Res 174:114–121. https://pubmed.ncbi.nlm.nih.gov/31055169/, https://doi.org/10.1016/j.envres.2019.04.001.
44.
Sontag-Padilla L, Burns RM, Shih RA, et al. The Urban Child Institute CANDLE Study: Methodological Overview and Baseline Sample Description. https://www.rand.org/pubs/research_reports/RR1336.html [accessed 30 May 2021].
45.
Barrett ES, Sathyanarayana S, Janssen S, Redmon JB, Nguyen RHN, Kobrosly R, et al. 2014. Environmental health attitudes and behaviors: findings from a large pregnancy cohort study. Eur J Obstet Gynecol Reprod Biol 176:119–125. https://pubmed.ncbi.nlm.nih.gov/24647207/, https://doi.org/10.1016/j.ejogrb.2014.02.029.
46.
Biederman J, Faraone SV, Doyle A, Lehman BK, Kraus I, Perrin J, et al. 1993. Convergence of the child behavior checklist with structured interview-based psychiatric diagnoses of ADHD children with and without comorbidity. J Child Psychol Psychiatry 34(7):1241–1251. https://pubmed.ncbi.nlm.nih.gov/8245144/, https://doi.org/10.1111/j.1469-7610.1993.tb01785.x.
47.
Chang LY, Wang MY, Tsai PS. 2016. Diagnostic accuracy of rating scales for attention-deficit/hyperactivity disorder: a meta-analysis. Pediatrics 137(3):e20152749. https://pubmed.ncbi.nlm.nih.gov/26928969/, https://doi.org/10.1542/peds.2015-2749.
48.
Achenbach TM, Rescorla LA. 2000. Manual for the ASEBA Preschool Forms and Profiles. Burlington, VT: University of Vermont, Research Center for Children, Youth, & Families.
49.
Achenbach TM, Rescorla L. 2001. Manual for the ASEBA School-Age Forms & Profiles. Burlington, VT: University of Vermont, Research Center for Children, Youth, & Families.
50.
Smucker MR, Craighead WE, Craighead LW, Green BJ. 1986. Normative and reliability data for the Children’s Depression Inventory. J Abnorm Child Psychol 14(1):25–39. https://pubmed.ncbi.nlm.nih.gov/3950219/, https://doi.org/10.1007/BF00917219.
51.
DiStefano C, Dombrowski SC. 2006. Investigating the theoretical structure of the Stanford-Binet-Fifth edition. J Psychoeduc Assess 24(2):123–136, https://doi.org/10.1177/0734282905285244.
52.
Roid GH, Pomplun M. 2012. The Stanford-Binet Intelligence Scales, Fifth Edition. In: Contemporary Intellectual Assessment: Theories, Tests, and Issues. 3rd ed. New York, NY: Guilford Press, 249–268.
53.
Rosenthal EN, Riccio CA, Gsanger KM, Jarratt KP. 2006. Digit span components as predictors of attention problems and executive functioning in children. Arch Clin Neuropsychol 21(2):131–139. https://pubmed.ncbi.nlm.nih.gov/16198530/, https://doi.org/10.1016/j.acn.2005.08.004.
54.
Wechsler D. WISC-V: Wechsler Intelligence Scale for Children. 2014. Bloomington, MN: Pearson.
55.
Sattler JM. 2018. Assessment of Children: Cognitive Foundations and Applications. 6th ed. La Mesa, CA: Jerome M. Sattler, Publisher, Inc.
56.
Wechsler D. 1967. WPPSI Manual: Wechsler Preschool and Primary Scale of Intelligence. San Antonio, TX: Psychological Corporation.
57.
Jelenkovic A, Mikkonen J, Martikainen P, Latvala A, Yokoyama Y, Sund R, et al. 2018. Association between birth weight and educational attainment: an individual-based pooled analysis of nine twin cohorts. J Epidemiol Community Health 72(9):832–837. https://pubmed.ncbi.nlm.nih.gov/29848580/, https://doi.org/10.1136/jech-2017-210403.
58.
Kullar SS, Shao K, Surette C, Foucher D, Mergler D, Cormier P, et al. 2019. A benchmark concentration analysis for manganese in drinking water and IQ deficits in children. Environ Int 130:104889. https://pubmed.ncbi.nlm.nih.gov/31200154/, https://doi.org/10.1016/j.envint.2019.05.083.
59.
Lanphear BP, Hornung R, Khoury J, Yolton K, Baghurst P, Bellinger DC, et al. 2005. Low-level environmental lead exposure and children’s intellectual function: an international pooled analysis. Environ Health Perspect 113(7):894–899. https://pubmed.ncbi.nlm.nih.gov/16002379/, https://doi.org/10.1289/ehp.7688.
60.
Wechsler D. 2003. Wechsler Intelligence Scale for Children Fourth Edition Response Book 1. 4th ed. San Antonio, TX: Psychological Corporation.
61.
Garred M, Gilmore L. 2009. To WPPSI or to Binet, that is the question: a comparison of the WPPSI-III and SB5 with typically developing preschoolers. Aust J Guid Couns 19(2):104–115, https://doi.org/10.1375/ajgc.19.2.104.
62.
Keller JP, Olives C, Kim S-Y, Sheppard L, Sampson PD, Szpiro AA, et al. 2015. A unified spatiotemporal modeling approach for predicting concentrations of multiple air pollutants in the Multi-Ethnic Study of Atherosclerosis and Air Pollution. Environ Health Perspect 123(4):301–309. https://pubmed.ncbi.nlm.nih.gov/25398188/, https://doi.org/10.1289/ehp.1408145.
63.
Kirwa K, Szpiro AA, Sheppard L, Sampson PD, Wang M, Keller JP, et al. 2021. Fine-Scale air pollution models for epidemiologic research: insights from approaches developed in the Multi-Ethnic Study of Atherosclerosis and Air Pollution (MESA Air). Curr Environ Health Rep 8(2):113–126. https://pubmed.ncbi.nlm.nih.gov/34086258/, https://doi.org/10.1007/s40572-021-00310-y.
64.
U.S. Bureau of Economic Analysis. Real Personal Income and Regional Price Parities. https://www.bea.gov/resources/methodologies/rpp [accessed 24 March 2022].
65.
Wechsler D. 1999. Wechsler Abbreviated Scale of Intelligence Record Form. Bloomington, MN: Pearson.
66.
McCrimmon AW, Smith AD. 2013. Review of the Wechsler Abbreviated Scale of Intelligence, second edition (WASI-II). J Psychoeduc Assess 31(3):337–341, https://doi.org/10.1177/0734282912467756.
67.
Wechsler D. 2011. Wechsler Abbreviated Scale of Intelligence. 2nd ed. Bloomington, MN: Pearson.
68.
Radloff LS. 1977. The CES-D scale: a Self-Report depression scale for research in the general population. Applied Psychological Measurement 1(3):385–401, https://doi.org/10.1177/014662167700100306.
69.
Pilkonis PA, Choi SW, Reise SP, Stover AM, Riley WT, Cella D, et al. PROMIS Cooperative Group. 2011. Item banks for measuring emotional distress from the Patient-Reported Outcomes Measurement Information System (PROMIS®): depression, anxiety, and anger. Assessment 18(3):263–283. https://pubmed.ncbi.nlm.nih.gov/21697139/, https://doi.org/10.1177/1073191111411667.
70.
Acevedo-Garcia D, McArdle N, Hardy EF, Crisan UI, Romano B, Norris D, et al. 2014. The child opportunity index: improving collaboration between community development and public health. Health Aff (Millwood) 33(11):1948–1957. https://pubmed.ncbi.nlm.nih.gov/25367989/, https://doi.org/10.1377/hlthaff.2014.0679.
71.
Benmarhnia T, Hajat A, Kaufman JS. 2021. Inferential challenges when assessing racial/ethnic health disparities in environmental research. Environ Health 20(1):7. https://pubmed.ncbi.nlm.nih.gov/33430882/, https://doi.org/10.1186/s12940-020-00689-5.
72.
Organisation for Economic Co-operation and Development Project on Income Distribution and Poverty. What are equivalence scales? https://www.oecd.org/els/soc/OECD-Note-EquivalenceScales.pdf [accessed 1 December 2021].
73.
Fisher RA. 2017. Statistical Methods for Research Workers. Gyan Books: New Delhi.
74.
VanderWeele TJ. 2019. Principles of confounder selection. Eur J Epidemiol 34(3):211–219. https://pubmed.ncbi.nlm.nih.gov/30840181/, https://doi.org/10.1007/s10654-019-00494-6.
75.
Hughes RA, Heron J, Sterne JAC, Tilling K. 2019. Accounting for missing data in statistical analyses: multiple imputation is not always the answer. Int J Epidemiol 48(4):1294–1304. https://pubmed.ncbi.nlm.nih.gov/30879056/, https://doi.org/10.1093/ije/dyz032.
76.
Kohavi R. 2001. A Study of Cross-Validation and Bootstrap for Accuracy Estimation and Model Selection. In: Proceedings of the 14th International Joint Conference on Artificial Intelligence – Vol. 2. 20–25 August 1995. Montreal, Quebec, Canada.
77.
Shapiro DE. 1999. The interpretation of diagnostic tests. Stat Methods Med Res 8(2):113–134. https://pubmed.ncbi.nlm.nih.gov/10501649/, https://doi.org/10.1177/096228029900800203.
78.
Azur MJ, Stuart EA, Frangakis C, Leaf PJ. 2011. Multiple imputation by chained equations: what is it and how does it work? Int J Methods Psychiatr Res 20(1):40–49. https://pubmed.ncbi.nlm.nih.gov/21499542/, https://doi.org/10.1002/mpr.329.
79.
Little RJA, Rubin DB. 2019. Statistical Analysis with Missing Data. Hoboken, NJ: John Wiley & Sons.
80.
Sejer EPF, Bruun FJ, Slavensky JA, Mortensen EL, Schiøler Kesmodel U. 2019. Impact of gestational age on child intelligence, attention and executive function at age 5: a cohort study. BMJ Open 9(9):e028982. https://pubmed.ncbi.nlm.nih.gov/31501108/, https://doi.org/10.1136/bmjopen-2019-028982.
81.
Palou-Serra A, Murcia M, Lopez-Espinosa M-J, Grimalt JO, Rodríguez-Farré E, Ballester F, et al. 2014. Influence of prenatal exposure to environmental pollutants on human cord blood levels of glutamate. Neurotoxicology 40:102–110. https://pubmed.ncbi.nlm.nih.gov/24361731/, https://doi.org/10.1016/j.neuro.2013.12.003.
82.
Marsit CJ. 2015. Influence of environmental exposure on human epigenetic regulation. J Exp Biol 218(pt 1):71–79. https://pubmed.ncbi.nlm.nih.gov/25568453/, https://doi.org/10.1242/jeb.106971.
83.
Basagaña X, Esnaola M, Rivas I, Amato F, Alvarez-Pedrerol M, Forns J, et al. 2016. Neurodevelopmental deceleration by urban fine particles from different emission sources: a longitudinal observational study. Environ Health Perspect 124(10):1630–1636. https://pubmed.ncbi.nlm.nih.gov/27128166/, https://doi.org/10.1289/EHP209.
84.
Guxens M, Garcia-Esteban R, Giorgis-Allemand L, Forns J, Badaloni C, Ballester F, et al. 2014. Air pollution during pregnancy and childhood cognitive and psychomotor development: six European birth cohorts. Epidemiology 25(5):636–647. https://pubmed.ncbi.nlm.nih.gov/25036432/, https://doi.org/10.1097/EDE.0000000000000133.
85.
Freire C, Ramos R, Puertas R, Lopez-Espinosa M-J, Julvez J, Aguilera I, et al. 2010. Association of traffic-related air pollution with cognitive development in children. J Epidemiol Community Health 64(3):223–228. https://pubmed.ncbi.nlm.nih.gov/19679705/, https://doi.org/10.1136/jech.2008.084574.
86.
Porta D, Narduzzi S, Badaloni C, Bucci S, Cesaroni G, Colelli V, et al. 2016. Air pollution and cognitive development at age 7 in a prospective Italian birth cohort. Epidemiology 27(2):228–236. https://pubmed.ncbi.nlm.nih.gov/26426942/, https://doi.org/10.1097/EDE.0000000000000405.
87.
Sentís A, Sunyer J, Dalmau-Bueno A, Andiarena A, Ballester F, Cirach M, et al. INMA Project. 2017. Prenatal and postnatal exposure to NO2 and child attentional function at 4-5 years of age. Environ Int 106:170–177. https://pubmed.ncbi.nlm.nih.gov/28689118/, https://doi.org/10.1016/j.envint.2017.05.021.
88.
Hazlehurst MF, Carroll KN, Loftus CT, et al. 2021. Maternal exposure to PM2.5 during pregnancy and asthma risk in early childhood: consideration of phases of fetal lung development. Environ Epidemiol 5(2):e130. https://pubmed.ncbi.nlm.nih.gov/33709049/, https://doi.org/10.1097/ee9.0000000000000130.
89.
Raz R, Kioumourtzoglou MA, Weisskopf MG. 2018. Live-birth bias and observed associations between air pollution and autism. Am J Epidemiol 187(11):2292–2296. https://pubmed.ncbi.nlm.nih.gov/30099488/, https://doi.org/10.1093/aje/kwy172.
90.
Harris MH, Gold DR, Rifas-Shiman SL, Melly SJ, Zanobetti A, Coull BA, et al. 2016. Prenatal and childhood traffic-related air pollution exposure and childhood executive function and behavior. Neurotoxicol Teratol 57:60–70. https://pubmed.ncbi.nlm.nih.gov/27350569/, https://doi.org/10.1016/j.ntt.2016.06.008.
91.
Adair-Rohani H. 2018. Air Pollution and Child Health: Prescribing Clean Air. Geneva, Switzerland: World Health Organization.
92.
Tulve N, Ruiz J, Lichtveld K, Darney S, Quackenboss J. 2016. Development of a conceptual framework depicting a child’s total (built, natural, social) environment in order to optimize health and well-being. J Environ Health Sci 2(2):1–8, https://doi.org/10.15436/2378-6841.16.1121.
93.
Konkel L. 2018. The brain before birth: using fMRI to explore the secrets of fetal neurodevelopment. Environ Health Perspect 126(11):112001. https://pubmed.ncbi.nlm.nih.gov/30457876/, https://doi.org/10.1289/EHP2268.
94.
Courchesne E, Chisum HJ, Townsend J, Cowles A, Covington J, Egaas B, et al. 2000. Normal brain development and aging: quantitative analysis at in vivo MR imaging in healthy volunteers. Radiology 216(3):672–682. https://pubmed.ncbi.nlm.nih.gov/10966694/, https://doi.org/10.1148/radiology.216.3.r00au37672.
95.
Giedd JN, Raznahan A, Alexander-Bloch A, Schmitt E, Gogtay N, Rapoport JL. 2015. Child psychiatry branch of the National Institute of Mental Health longitudinal structural magnetic resonance imaging study of human brain development. Neuropsychopharmacology 40(1):43–49, https://doi.org/10.1038/npp.2014.236.
96.
Kennedy DN, Makris N, Herbert MR, Takahashi T, Caviness VS. 2002. Basic principles of MRI and morphometry studies of human brain development. Dev Sci 5(3):268–278, https://doi.org/10.1111/1467-7687.00366.
97.
Durston S, Hulshoff Pol HE, Casey BJ, Giedd JN, Buitelaar JK, van Engeland H. 2001. Anatomical MRI of the developing human brain: what have we learned? J Am Acad Child Adolesc Psychiatry 40(9):1012–1020. https://pubmed.ncbi.nlm.nih.gov/11556624/, https://doi.org/10.1097/00004583-200109000-00009.
98.
Calderón-Garcidueñas L, Mora-Tiscareño A, Ontiveros E, Gómez-Garza G, Barragán-Mejía G, Broadway J, et al. 2008. Air pollution, cognitive deficits and brain abnormalities: a pilot study with children and dogs. Brain Cogn 68(2):117–127. https://pubmed.ncbi.nlm.nih.gov/18550243/, https://doi.org/10.1016/j.bandc.2008.04.008.
99.
Anderson P. 2002. Assessment and development of executive function (EF) during childhood. Child Neuropsychol 8(2):71–82. https://pubmed.ncbi.nlm.nih.gov/12638061/, https://doi.org/10.1076/chin.8.2.71.8724.
100.
Calderón-Garcidueñas L, Azzarelli B, Acuna H, Garcia R, Gambling TM, Osnaya N, et al. 2002. Air pollution and brain damage. Toxicol Pathol 30(3):373–389. https://pubmed.ncbi.nlm.nih.gov/12051555/, https://doi.org/10.1080/01926230252929954.
101.
Campbell A, Oldham M, Becaria A, Bondy SC, Meacher D, Sioutas C, et al. 2005. Particulate matter in polluted air may increase biomarkers of inflammation in mouse brain. Neurotoxicology 26(1):133–140. https://pubmed.ncbi.nlm.nih.gov/15527881/, https://doi.org/10.1016/j.neuro.2004.08.003.
102.
Semmler A, Okulla T, Sastre M, Dumitrescu-Ozimek L, Heneka MT. 2005. Systemic inflammation induces apoptosis with variable vulnerability of different brain regions. J Chem Neuroanat 30(2–3):144–157. https://pubmed.ncbi.nlm.nih.gov/16122904/, https://doi.org/10.1016/j.jchemneu.2005.07.003.
103.
Thomason ME, Race E, Burrows B, Whitfield-Gabrieli S, Glover GH, Gabrieli JDE. 2009. Development of spatial and verbal working memory capacity in the human brain. J Cogn Neurosci 21(2):316–332. https://pubmed.ncbi.nlm.nih.gov/18510448/, https://doi.org/10.1162/jocn.2008.21028.
104.
Stingone JA, McVeigh KH, Claudio L. 2016. Association between prenatal exposure to ambient diesel particulate matter and perchloroethylene with children’s 3rd grade standardized test scores. Environ Res 148:144–153. https://pubmed.ncbi.nlm.nih.gov/27058443/, https://doi.org/10.1016/j.envres.2016.03.035.
105.
Curtis JT, Hood AN, Chen Y, Cobb GP, Wallace DR. 2010. Chronic metals ingestion by prairie voles produces sex-specific deficits in social behavior: an animal model of autism. Behav Brain Res 213(1):42–49. https://pubmed.ncbi.nlm.nih.gov/20433873/, https://doi.org/10.1016/j.bbr.2010.04.028.
106.
Spritzer MD, Galea LAM. 2007. Testosterone and dihydrotestosterone, but not estradiol, enhance survival of new hippocampal neurons in adult male rats. Dev Neurobiol 67(10):1321–1333. https://pubmed.ncbi.nlm.nih.gov/17638384/, https://doi.org/10.1002/dneu.20457.
107.
Hajat A, MacLehose RF, Rosofsky A, Walker KD, Clougherty JE. 2021. Confounding by socioeconomic status in epidemiological studies of air pollution and health: challenges and opportunities. Environ Health Perspect 129(6):65001. https://pubmed.ncbi.nlm.nih.gov/34124937/, https://doi.org/10.1289/EHP7980.
108.
Wesselhoeft R, Davidsen K, Sibbersen C, Kyhl H, Talati A, Andersen MS, et al. 2021. Maternal prenatal stress and postnatal depressive symptoms: discrepancy between mother and teacher reports of toddler psychological problems. Soc Psychiatry Psychiatr Epidemiol 56(4):559–570. https://pubmed.ncbi.nlm.nih.gov/32995941/, https://doi.org/10.1007/s00127-020-01964-z.
109.
Olino TM, Michelini G, Mennies RJ, Kotov R, Klein DN. 2021. Does maternal psychopathology bias reports of offspring symptoms? A study using moderated non-linear factor analysis. J Child Psychol Psychiatry 62(10):1195–1201. https://pubmed.ncbi.nlm.nih.gov/33638150/, https://doi.org/10.1111/jcpp.13394.
110.
Spiegelman D. 2010. Approaches to uncertainty in exposure assessment in environmental epidemiology. Annu Rev Public Health 31:149–163. https://pubmed.ncbi.nlm.nih.gov/20070202/, https://doi.org/10.1146/annurev.publhealth.012809.103720.
111.
Szpiro AA, Paciorek CJ. 2013. Measurement error in two-stage analyses, with application to air pollution epidemiology. Environmetrics 24(8):501–517. https://pubmed.ncbi.nlm.nih.gov/24764691/, https://doi.org/10.1002/env.2233.
112.
Alwan S, Chambers CD. 2015. Identifying human teratogens: an update. J Pediatr Genet 4(2):39–41. https://pubmed.ncbi.nlm.nih.gov/27617116/, https://doi.org/10.1055/s-0035-1556745.
113.
Simpson JL. 1990. Incidence and timing of pregnancy losses: relevance to evaluating safety of early prenatal diagnosis. Am J Med Genet 35(2):165–173. https://pubmed.ncbi.nlm.nih.gov/2178414/, https://doi.org/10.1002/ajmg.1320350205.
114.
Cunningham FG, MacDonald PC, Gant NF, Leveno KJ, Gilstrap LC. 1993. Williams Obstetrics 19th Edition. New York, NY: Appleton & Lange.
115.
Sawin SW, Morgan MA. 1996. Dating of pregnancy by trimesters: a review and reappraisal. Obstet Gynecol Surv 51(4):261–264. https://pubmed.ncbi.nlm.nih.gov/8657403/, https://doi.org/10.1097/00006254-199604000-00023.
116.
Basagaña X, Barrera-Gómez J. 2022. Reflection on modern methods: visualizing the effects of collinearity in distributed lag models. Int J Epidemiol 51(1):334–344. https://pubmed.ncbi.nlm.nih.gov/34458914/, https://doi.org/10.1093/ije/dyab179.
117.
Perkins NJ, Cole SR, Harel O, Tchetgen EJ, Sun B, Mitchell EM, et al. 2018. Principled approaches to missing data in epidemiologic studies. Am J Epidemiol 187(3):568–575. https://pubmed.ncbi.nlm.nih.gov/29165572/, https://doi.org/10.1093/aje/kwx348.
118.
Clark C, Crombie R, Head J, van Kamp I, van Kempen E, Stansfeld SA. 2012. Does traffic-related air pollution explain associations of aircraft and road traffic noise exposure on children’s health and cognition? A secondary analysis of the United Kingdom sample from the RANCH project. Am J Epidemiol 176(4):327–337. https://pubmed.ncbi.nlm.nih.gov/22842719/, https://doi.org/10.1093/aje/kws012.

Information & Authors

Information

Published In

Environmental Health Perspectives
Volume 130Issue 6June 2022
PubMed: 35737514

History

Received: 2 September 2021
Revision received: 26 May 2022
Accepted: 6 June 2022
Published online: 23 June 2022

Authors

Affiliations

Yu Ni
Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, Washington, USA
Christine T. Loftus
Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, Washington, USA
Adam A. Szpiro
Department of Biostatistics, School of Public Health, University of Washington, Seattle, Washington, USA
Michael T. Young
Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, Washington, USA
Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, Washington, USA
Laura E. Murphy
Department of Psychiatry, University of Tennessee Health Science Center, Memphis, Tennessee, USA
Frances A. Tylavsky
Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
W. Alex Mason
Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
Kaja Z. LeWinn
Department of Psychiatry, School of Medicine, University of California, San Francisco, San Francisco, California, USA
Sheela Sathyanarayana
Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, Washington, USA
Department of Pediatrics, School of Medicine, University of Washington, Seattle, Washington, USA
Seattle Children’s Research Institute, Seattle, Washington, USA
Emily S. Barrett
Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, New Jersey, USA
Department of Psychiatry, School of Medicine, University of California, San Francisco, San Francisco, California, USA
Department of Pediatrics, School of Medicine, University of California, San Francisco, San Francisco, California, USA
Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, Washington, USA
Department of Pediatrics, School of Medicine, University of Washington, Seattle, Washington, USA
Department of Epidemiology, School of Public Health, University of Washington, Seattle, Washington, USA

Notes

Address correspondence to Yu Ni, 4225 Roosevelt Way NE, Rm. 301, Seattle, WA 98105 USA. Email: [email protected]

Metrics & Citations

Metrics

About Article Metrics


Citations

Download citation

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Simply select your manager software from the list below and click DOWNLOAD.

Cited by

  • Susceptible windows of prenatal and postnatal fine particulate matter exposures and attention-deficit hyperactivity disorder symptoms in early childhood, Science of The Total Environment, 10.1016/j.scitotenv.2023.168806, 912, (168806), (2024).
  • Sex-specific associations between placental corticotropin releasing hormone and problem behaviors in childhood, Psychoneuroendocrinology, 10.1016/j.psyneuen.2024.106994, 163, (106994), (2024).
  • What can be done to protect toddlers from air pollution: Current evidence, Journal of Pediatric Nursing, 10.1016/j.pedn.2024.01.018, (2024).
  • Relation of prenatal and postnatal PM2.5 exposure with cognitive and motor function among preschool-aged children, International Journal of Hygiene and Environmental Health, 10.1016/j.ijheh.2023.114317, 256, (114317), (2024).
  • Associations between maternal exposure to air pollution during pregnancy and trajectories of infant growth: A birth cohort study, Ecotoxicology and Environmental Safety, 10.1016/j.ecoenv.2023.115792, 269, (115792), (2024).
  • School Bus Rebate Program and Student Educational Performance Test Scores, JAMA Network Open, 10.1001/jamanetworkopen.2024.3121, 7, 3, (e243121), (2024).
  • Association between maternal occupational exposure to cleaning chemicals during pregnancy and childhood wheeze and asthma, Frontiers in Epidemiology, 10.3389/fepid.2023.1166174, 3, (2023).
  • Associations of Exposure to Air Pollution during the Male Programming Window and Mini-Puberty with Anogenital Distance and Penile Width at Birth and at 1 Year of Age in the Multicenter U.S. TIDES Cohort, Environmental Health Perspectives, 10.1289/EHP12627, 131, 11, (2023).
  • undefined, 2023 11th International Symposium on Digital Forensics and Security (ISDFS), 10.1109/ISDFS58141.2023.10131695, (1-6), (2023).
  • Wildfire smoke, environmental justice, and young children in urban Pacific Northwest communities, Urban Climate, 10.1016/j.uclim.2023.101581, 50, (101581), (2023).
  • See more

View Options

View options

PDF

View PDF

Get Access

Restore your content access

Enter your email address to restore your content access:

Note: This functionality works only for purchases done as a guest. If you already have an account, log in to access the content to which you are entitled.

Media

Figures

Other

Tables

Share

Share

Copy the content Link

Share on social media