Abstract
Background:
Maternal smoking during pregnancy is related to altered DNA methylation in infant umbilical cord blood. The extent to which low levels of smoke exposure among nonsmoking pregnant women relates to offspring DNA methylation is unknown.
Objective:
This study sought to evaluate relationships between maternal prenatal plasma cotinine levels and DNA methylation in umbilical cord blood in newborns using the Infinium HumanMethylation 450K BeadChip.
Methods:
Participants from the Newborn Epigenetics Study cohort who reported not smoking during pregnancy had verified low levels of cotinine from maternal prenatal plasma (0 to ), and offspring epigenetic data from umbilical cord blood were included in this study (). Multivariable linear regression models were fit to the data, controlling for cell proportions, age, race, education, and parity. Estimates represent changes in response to any unit increase in exposure.
Results:
Multivariable linear regression models yielded 29,049 CpGs that were differentially methylated in relation to increases in cotinine at a 5% false discovery rate. Top CpGs were within or near genes involved in neuronal functioning (PRKG1, DLGAP2, BSG), carcinogenesis (FHIT, HSPC157) and inflammation (AGER). Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses suggest cotinine was related to methylation of gene pathways controlling neuronal signaling, metabolic regulation, cell signaling and regulation, and cancer. Further, enhancers associated with transcription start sites were enriched in altered CpGs. Using an independent sample from the same study population (), bisulfite pyrosequencing was performed with infant cord blood DNA for two genes within our top 20 hits (AGER and PRKG1). Results from pyrosequencing replicated epigenome results for PRKG1 (cg17079497, , , ) but not for AGER (cg09199225; , , ).
Discussion:
Secondhand smoke exposure among nonsmoking women may alter DNA methylation in regions involved in development, carcinogenesis, and neuronal functioning. These novel findings suggest that even low levels of smoke exposure during pregnancy may be sufficient to alter DNA methylation in distinct sites of mixed umbilical cord blood leukocytes in pathways that are known to be altered in cord blood from pregnant active smokers. https://doi.org/10.1289/EHP8099
Introduction
Smoking is consistently linked with alterations in DNA methylation (Philibert et al. 2012, 2013; Wan et al. 2012; Zeilinger et al. 2013). Several epigenome-wide association studies (EWAS) comparing smokers and nonsmokers demonstrate that self-reported smoking is associated with alterations in DNA methylation at multiple cytosine-phosphate-guanine (CpG) dinucleotide sites that can lead to changes in gene transcription (Dogan et al. 2014; Elliott et al. 2014; Shenker et al. 2013; Zaghlool et al. 2015; Zhang et al. 2016; Zhu et al. 2016). There is also evidence to suggest that alterations in DNA methylation can be induced even when people have smoked for only a short time (Philibert et al. 2012, 2013; Prince et al. 2019).
Tobacco smoke exposure in utero has also been associated with alterations in DNA methylation across many genes affecting different tissue types (Joubert et al. 2012; Richmond et al. 2015; Suter et al. 2013; Suter and Aagaard 2012). For example, global hypomethylation has been detected in buccal cells and peripheral blood granulocytes of children exposed to prenatal smoking (Breton et al. 2009). Some altered methylated markers have been found to persist over time in the offspring epigenome (Richmond et al. 2015; Wiklund et al. 2019), with studies reporting evidence of persistence across the lifespan from childhood and adolescence (Lee et al. 2015; Richmond et al. 2015) to adulthood (Richmond et al. 2018; Tehranifar et al. 2018).
Smoking-related alterations in DNA methylation have been frequently studied using the umbilical cord blood samples of children exposed to smoke prenatally (Bergens et al. 2019; Ivorra et al. 2015; Miyake et al. 2018; Zhang et al. 2018). The largest meta-analyses of the association between maternal smoking during pregnancy and DNA methylation of newborn umbilical cord blood at over 450,000 CpG sites was conducted by Joubert et al. (2016). Data were meta-analyzed across the Pregnancy And Childhood Epigenetics consortium, which includes data from 13 cohorts (). More than 6,000 CpGs were differentially methylated in relation to self-reported maternal smoking, dichotomized as smokers vs. nonsmokers, including 2,965 CpGs corresponding to 2,017 genes not previously related to smoking and methylation in children (Joubert et al. 2016). The top hit was aryl-hydrocarbon receptor repressor (AHRR) cg05575921, which has been observed previously as differentially methylated in relation to active smoking in adults and secondhand smoke exposure in children (Joubert et al. 2012, 2016; Monick et al. 2012; Shenker et al. 2013; Zeilinger et al. 2013). Differential DNA methylation has also been reported, within Myosin IG (MY01G), Growth Factor Independent 1 Transcriptional Repressor (GFI1), and CYP1A1 (Breitling et al. 2011; Joubert et al. 2012; Kirchner et al. 2013; Monick et al. 2012; Shenker et al. 2013). These loci have been implicated in susceptibility to orofacial clefts, tooth development and eruption, asthma, hepatocellular carcinoma, and colorectal and breast cancers (Joubert et al. 2016).
These studies are highly informative to our understanding of the potential consequences of maternal smoking during pregnancy. However, exposure to secondhand smoke during pregnancy among nonsmokers is more common than active smoking during pregnancy. Using data from a U.S. nationally representative study, the Population Assessment of Tobacco and Health Study (2013–2015), our group found that 23% of pregnant women (ages 18–54 y) reported exposure to secondhand smoke, whereas only 6.1% reported smoking during pregnancy (Do et al. 2018). Although the adverse health outcomes associated with secondhand smoke exposure and active smoking during pregnancy are similar for mothers and newborns (Centers for Disease Control and Prevention 2020), the epigenetic consequences on the newborn epigenome of secondhand smoke exposure among nonsmoking women is not known. The reason for this may be the difficulty in assessing secondhand smoke exposure among nonsmoking women. Studies of active smoking during pregnancy have relied on self-report, but assessing secondhand smoke exposure by self-report can be a challenge. There is a risk for bias in self-report measures, especially among pregnant women who are either unaware of their levels of exposure or, because of social desirability, underreport their levels of exposure (Garg et al. 2016; Schechter et al. 2018). A more accurate method to assess secondhand smoke exposure among pregnant women is the use of biomarkers, such as cotinine, a metabolite of nicotine (Philibert et al. 2013).
To our knowledge, no published studies have examined alterations in DNA methylation in infant cord blood as it relates to secondhand smoke exposure during pregnancy. However, there is evidence of associations between secondhand smoke exposure and alterations in DNA methylation in adults from the Multi-Ethnic Study of Atherosclerosis study (Reynolds et al. 2017), as well as experimental evidence of that association in mice (Noël et al. 2017). Knowledge of the DNA methylation loci that may be altered by prenatal secondhand smoke exposure could help identify biomarkers of exposure when maternal cotinine is not available. Equally important to public health is knowing to what extent DNA methylation is altered among nonsmoking women who are exposed to tobacco smoke in their everyday environment. To address these gaps, we conducted an EWAS study to investigate alterations in DNA methylation among a sample of newborns born to nonsmoking pregnant mothers and performed pyrosequencing on select loci in an independent sample from the same cohort to replicate some of our EWAS findings. The results strengthen the case for continued clinical and policy interventions to mitigate any level of smoke exposure during pregnancy, because the findings here appear to suggest that variation, even at lower levels consistent with secondhand smoke exposure, may have the potential to affect the epigenome.
Materials and Methods
Study Population
Participants included in the current analyses are a part of the Newborn Epigenetic STudy (NEST), an ongoing prebirth cohort study designed to improve our understanding of the environmental influences on epigenetic responses and phenotypes in children (Hoyo et al. 2011; Liu et al. 2012). Study participants were identified among pregnant women attending Duke University-affiliated prenatal clinics in Durham, North Carolina, between 2005 and 2011. To be eligible for the NEST study, participants had to be at least 18 years of age or older, English or Spanish speaking, planning to use Duke or Durham Regional Hospital for delivery for the index pregnancy, and willing to provide a prenatal blood sample. Exclusion criteria included women intending to move before the first birthday of the offspring, relinquish custody of the index child, or who had confirmed human immunodeficiency virus (HIV) infection among the first third of the cohort only.
A total of 2,681 mother–child pairs were enrolled and consented. Information on covariates (i.e., race/ethnicity, maternal education, and maternal smoking during pregnancy) was ascertained during the enrollment survey, whereas mother’s age at delivery and parity were ascertained through medical records. DNA methylation analyses were completed for 427 with adequate infant umbilical cord blood samples and a minimum amount of follow-up data. Cotinine was assayed from prenatal maternal plasma samples among mother who had singleton births and who had agreed to allow their samples to be used in future research. These cases formed the basis for the analytic samples included in the 450K Beadchip and pyrosequencing analyses, as further described below.
The analytical sample for the 450K Beadchip analyses () was restricted to those who reported their race/ethnicity as non-Hispanic White or Black and those for whom we had completed cotinine assays from maternal prenatal plasma. In addition, cotinine values had to be less than , a threshold proposed by Benowitz et al. as being consistent with secondhand smoke exposure in the U.S. population (Benowitz et al. 2009). Offspring eligibility requirements were limited to live births and singletons. There were no requirements regarding the child’s health at birth.
The analytical sample used for validation using pyrosequencing was restricted to those who were not included in 450K Beadchip analyses, those who reported their race/ethnicity as non-Hispanic White or Black, those with cotinine levels lower than , and those who had data on necessary covariates for analysis (). Covariates included race/ethnicity (categorical variable, with responses being: Black, non-Hispanic White), mother’s age at delivery (continuous variable, reported in years), maternal education [categorical variable, with responses being: less than high school, high school diploma or general education diploma (GED), some college, or college graduate], and parity (categorical variable, with responses being: 0, 1, 2, or 3 or more) for both 450K and pyrosequencing analyses and additional technical covariates (plate, batch) for the 450K Beadchip analyses.
Ethical Approval
The NEST has been approved by the institutional review board at Duke University (Pro00043781, Pro00014548, and Pro00064859) and Virginia Commonwealth University (HM20007857) and have been performed in accordance with the 1964 Helsinki Declaration and its later amendments. Written informed consent was provided by all participants in this study.
Data Collection Methods and Measures
Maternal prenatal cotinine collection and assay procedures.
Maternal blood (plasma) specimens were collected with survey data on health, nutrition, stress, and lifestyle behaviors during the enrollment period, which occurred during the first trimester [mean gestational , weeks] for most participants. As described in greater detail elsewhere (Schechter et al. 2018), assays were completed at the Exposure Biology and Chemistry Lab at Duke University. Cotinine was measured using high-performance liquid chromatography coupled with tandem mass spectrometric detection (HPLC-MS-MS) method. This highly sensitive assay was designed to measure levels of secondhand smoke exposure with a limit of detection (LOD) of and a reproducibility (Bernert et al. 2009; Dempsey et al. 2012; Jacob et al. 2011). We assumed the value of for all values under the LOD for these analyses. No mothers in the 450K Beadchip analyses and 3 mothers in the pyrosequencing analyses had values under LOD.
Assessment of maternal smoking based on self-report and cotinine values.
At the time the plasma sample was collected, mothers indicated by survey whether they had smoked at all during pregnancy. Mothers were classified as “nonsmokers” if they had indicated that they had not smoked at any point during their pregnancy. To ensure that our sample included only nonsmoking mothers, we combined the self-reported survey information with cotinine concentration values from prenatal maternal plasma. The resulting analytic sample included only self-reported nonsmoking mothers, who had cotinine concentration levels of , with nonzero levels indicative of secondhand smoke exposure according to thresholds proposed by Benowitz et al. (2009). Cotinine concentration values ranging from for maternal plasma were retained as the main exposure of interest for the statistical analyses and was treated as a continuous variable.
Offspring umbilical cord blood collection and DNA methylation.
Infant umbilical cord blood was collected via umbilical vein puncture into ethylenediaminetetraacetic acid (EDTA)-containing vacutainer tubes, inverted to mix, and centrifuged to harvest plasma and the leukocyte-containing buffy coat used for DNA extraction. Specimens were stored at until the time of analysis (Murphy et al. 2012). Offspring DNA was extracted using Puregene reagents (Qiagen) according to the manufacturer’s protocol. The resulting DNA was assessed for concentration and purity using a Thermo Scientific™ NanoDrop™ 2000 spectrophotometer (ThermoFisher Scientific). Offspring genomic DNA () was modified by treatment with sodium bisulfite using the Zymo EZ DNA Methylation Kit (Zymo Research). As explained elsewhere (Gonzalez-Nahm et al. 2018), bisulfite treatment of denatured DNA converts all unmethylated cytosines to uracils, leaving methylated cytosines unchanged and allowing for quantitative measurement of cytosine methylation status. Methylation levels for individual CpG sites were then measured using the Illumina Infinium® HumanMethylation450 BeadChip (hereafter, “450K Beadchip”; Illumina, Inc.) at the Duke Molecular Genomics Core Facility. The 450K BeadChip interrogates more than 480,000 methylation sites (Bibikova et al. 2011).
Pyrosequencing.
We performed bisulfite pyrosequencing using DNA from infant cord blood from a subsample of newborns from the NEST cohort who were not included in 450K BeadChip analyses. Cases were selected from all participants with infant cord blood and prenatal maternal plasma samples not included in 450K analyses and were intentionally selected to be similar to those included in the 450K Beadchip analyses across key maternal characteristics, specifically nonsmoking, Black or non-Hispanic White, and prenatal cotinine levels between 0 to . These cases were intentionally selected to be similar to those included in the 450K BeadChip analyses across maternal characteristics.
We assessed DNA methylation at two regions associated with genes within our top 20 hits based on smallest p-value demonstrating infant cord blood methylation differences in relation to cotinine concentration from prenatal maternal plasma (AGER and PRKG1). Pyrosequencing was performed using a PyroMark® Q96 MD pyrosequencer (Qiagen). Assays were designed using the PyroMark Assay Design Software (Qiagen). The Qiagen PyroMark® PCR Kit was used for amplification of the template, using of template DNA and of a stock of each forward and reverse primer in a reaction volume. Polymerase chain reaction (PCR) conditions were as follows: followed by 60 cycles of 94°C × 30 s, 61°C × 30 s, 72°C × 30 s; a final extension at 72°C followed by a 4°C hold. PCR primers for AGER were F: 5′-biotin-ATA TGT GAT TGG GGG GAT GGT-3′ and R: 5′-CCA CAA AAT AAC CCC AAT AAA CAA-3′ and the sequencing primer: 5′-CCT CCC ACA AAA CCT ATA-3′. The AGER sequence to analyze was 5′-CRA AAA CAA AAA AAA TTA AAA ACA CAA C-3′. The underlined CpG position (on the reverse complement strand) corresponds to 450K BeadChip probe cg09199225. For PRKG1, PCR primers were F: 5′-biotin-GGA GTT AAA TGG AGA AAG ATA AGG A-3′, R: 5′-CTC TTC CTC AAA ATC CTA CCT AAA T-3′ and the sequencing primer: 5′CTA AAA ACT CTA ATA CTT CA-3′. The PRKG1 sequence to analyze was 5′-AAT CA ACCT CTC TAA ACA ATT ACA CRC AAA AAA ACC CAC TCT TAA AAA AAT TTC TCC AAA ATC CTT ATC TTT CT-3, with the underlined CpG corresponding to 450K BeadChip probe cg17079497. Assay performance was verified using mixed unmethylated and methylated bisulfite controls (EpiTect DNA; Qiagen). Percent methylation for each CpG was determined using Pyro Q-CpG Software (Qiagen). Linear regression analyses, which included the same variables as covariates in the 450K BeadChip analysis, were performed in SAS (version 9.4; SAS Institute Inc.).
Statistical Analyses
Genome-Wide DNA methylation analysis.
To investigate the effect of secondhand smoke exposure among self-reported nonsmoking mothers on newborn DNA methylation, we performed analysis using DNA obtained from infant cord blood on the Infinium® 450K BeadChip. Analysis was performed in R (version 3.4.0; R Development Core Team)/Bioconductor (version 3.5; Bioconductor) environment. Probe intensity data (IDAT) files were processed using the RnBead (version 1.8.0) R package. Probes outside of CpG context, containing SNPs at any probe position, probes for the X and Y chromosomes, and low variability () probes were removed based on best practices recommendations (Chen et al. 2013b; Pidsley et al. 2016). Method wm.dasen was used for data normalization (Pidsley et al. 2016). Background correction was performed with method methylumi.noob (Davis et al. 2020). A matrix of values was obtained. Each value represents the proportion methylated for the beadtype on the array, which ranges from 0 (unmethylated) to 1 (completely methylated). Quality of each array was assessed by examining density plots of the values, bean plots of all values, and values for various control beads. A total of 385,265 probes were left for analyses following quality control procedures. The association between methylation and cotinine level was performed using beta regression as implemented in the betareg (version 3.1-0) R package, which explicitly accounted for the bimodal nature of methylation data (Cribari-Neto and Zeileis 2010). Beta regression has been shown to improve the detection of differential DNA methylation for epigenetic epidemiological studies in comparison to other approaches (Triche et al. 2016). The following covariates were included in the model: race/ethnicity, mother’s age at delivery, maternal education, parity, and technical covariates (plate, row, column). Houseman-estimated cell proportions (Houseman et al. 2012) with the Reinius et al. (2012) data set for reference (Reinius et al. 2012) were also included as fixed effects to correct for cell mixture distribution. The resulting p-values were corrected using Benjamini and Hochberg’s false discovery rate (FDR) method (Benjamini and Hochberg 1995). CpG sites with FDR were considered statistically significant.
Functional enrichment analysis.
Functional enrichment analysis was performed using EnrichR (Chen et al. 2013a). The results of Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis were visualized using the “pathview” R package (version 1.16.7). Given that there were a large number of genes () found to be significantly associated with alterations in the DNA methylation of infant cord blood, functional enrichment analyses were run on the top 3,000 genes most highly related to cotinine.
Epigenomic enrichment analysis.
As a follow-up to the KEGG functional enrichment analysis, epigenomic enrichment analyses were conducted. Specifically, GenomeRunner (Dozmorov et al. 2012, 2016) was used to test whether hg19 genomic coordinates of CpG sites associated with low-level secondhand smoke exposure (as measured by cotinine levels) were significantly enriched in certain genomic annotations collected by the ENCODE, Roadmap, and other consortia, as compared with randomly sampled CpGs from all CpGs on the 450K BeadChip. Briefly, hypergeometric tests were used to calculate enrichment/depletion at specific CpG sites, and p-values were FDR-corrected for multiple testing. Given that methylation profiles were obtained from blood samples, cell type-specific genome annotation data from Gm12878 cell line (B-cell lymphoblastoma) were used.
DNA methylation analysis at select gene regions by pyrosequencing.
Linear regression analyses were conducted to determine associations between CpG markers that were pyrosequenced within two select gene regions and alterations in DNA methylation of umbilical cord blood, including the same covariates in the 450K BeadChip analysis. These analyses were performed in SAS (version 9.4; SAS Institute).
Pyrosequencing assays for AGER and PRKG1 were designed to capture the select CpG sites that were significant for each on the 450K BeadChip platform. CpGs were selected from a larger set of CpGs that were significant in the 450K BeadChip sample, based on statistical significance, the magnitude of effect size, and the ability to design and validate pyrosequencing assays to capture the specific CpG sites represented in the 450K BeadChip platform. Assay design was first validated using defined mixtures of fully methylated and unmethylated bisulfite modified control DNA (0%, 25%, 50%, 75%, and 100% methylated DNA), which showed good agreement between the amount of methylated DNA in the reaction and what was measured by pyrosequencing (AGER: ; PKRG1: ). Due to the stringent internal quality controls that were imposed for this pyrosequencing reaction, more samples were excluded for AGER. We then used these assays to measure DNA methylation in umbilical cord blood for an independent subset of participants in the NEST cohort for whom 450K BeadChip data were not generated.
Results
Sample Characteristics
Sample characteristics and comparisons between individuals included in the 450K BeadChip and pyrosequencing analyses are displayed in Table 1. Of the 79 pregnant women included in the 450K Beadchip analysis, 45.6% () indicated that their race/ethnicity was Black, and 54.4% () indicated that their race/ethnicity was non-Hispanic White. Nearly half (, or 48.1%) of the sample were college graduates. For 35.4% () of the sample, the index pregnancy was their first pregnancy. The average age of mothers at delivery was 29.2 y (range: 18–45 y, y). Cotinine concentration in plasma from blood samples collected from pregnant women ranged from 0 to (, ; , range: ). None of the cotinine values were under the LOD. On average, mothers delivered their babies at 39.0 (; , range: 32.2–41.7) wk gestation.
Table 1.
Sample characteristics.
Full sample () | Included in 450K BeadChip analyses () | Included in pyrosequencing analyses () | |||||
---|---|---|---|---|---|---|---|
Variable | n (%) or | Median (range) | n (%) or | Median (range) | n (%) or | Median (range) | -Value |
Race/ethnicity | 0.01 | ||||||
Black | 1,166 (43.6) | 36 (45.6) | 75 (65.2) | ||||
Hispanic | 478 (17.9) | 0 (0.0) | 0 (0.0) | ||||
Other race/ethnicity | 125 (4.7) | 0 (0.0) | 0 (0.0) | ||||
Non-Hispanic White | 908 (33.9) | 43 (54.4) | 40 (34.8) | ||||
Education status | 0.33 | ||||||
Less than high school | 526 (21.6) | 3 (3.8) | 12 (10.6) | ||||
High school/GED | 556 (22.9) | 19 (24.1) | 22 (19.5) | ||||
Some college | 542 (22.3) | 19 (24.1) | 29 (25.7) | ||||
College graduate | 807 (33.2) | 38 (48.0) | 50 (44.3) | ||||
Parity | 0.31 | ||||||
0 | 829 (33.9) | 28 (35.4) | 48 (41.7) | ||||
1 | 788 (32.2) | 22 (27.9) | 35 (30.4) | ||||
2 | 483 (19.7) | 18 (22.8) | 23 (20.0) | ||||
3 or more | 347 (14.2) | 11 (13.9) | 9 (7.8) | ||||
Maternal smoking during pregnancy | Not computed | ||||||
No | 1,834 (75.9) | 79 (100.0) | 115 (100.0) | ||||
Yes | 582 (24.1) | 0 (0.0) | 0 (0.0) | ||||
Mother’s age at delivery (y) | 28.0 (18.0–40.0) | 30.0 (18.0–45.0) | 28.0 (18.0–40.0) | 0.20 | |||
Gestational age at birth (wk) | 39.1 (18.4–42.3) | 39.1 (32.2–41.7) | 39.3 (33.5–41.4) | 0.35 | |||
Gestational age at maternal plasma collection (wk) | 13.9 (3.3–42.0) | 38.9 (3.3–41.6) | 11.3 (5.3–40.9) | ||||
Cotinine concentration in maternal plasma (ng/mL) | 0.6 (0.0–371.0) | 1.0 (0.0–3.8) | 0.5 (0.0–3.6) |
Note. p-Values reflect comparisons between the 450K sample and the pyrosequencing sample only. The p-value for maternal smoking during pregnancy was not computed because both 450K and pyrosequencing analyses included mothers who did not smoke during pregnancy. GED, general education diploma; SD, standard deviation.
Of the 115 pregnant women included within the pyrosequencing sample, 65.2% () were Black, and 34.8% () were non-Hispanic White. Less than half (44.3%, ) were college graduates. The average age at delivery was 28.0 y (range: 18 to 40 y; ). Cotinine concentration in plasma from maternal blood samples ranged from 0 to (, ; , range: 0.0–3.6). Three of the cotinine values were under the LOD and assigned a value of . Mothers were on average 38.0 (; , range: 33.5–41.4) years of age when they delivered their babies.
There were no significant differences between those in the 450K BeadChip sample and those in the pyrosequencing sample except for race/ethnicity and cotinine values (Table 1). The percent of Black mothers was greater in the pyrosequencing sample than the 450K BeadChip sample (65.2% vs. 45.6%, ), and cotinine concentrations were statistically significantly more elevated in the 450K BeadChip analyses relative to those in the pyrosequencing sample (; ; ; range: vs. ; ; ; , ). In addition, the timing of the maternal prenatal plasma collection differed between the 450K BeadChip and pyrosequencing samples (mean weeks of gestational ; ; ; range: 3.3–41.6 vs. ; ; ; , ). Histograms of maternal cotinine in plasma (nanograms per milliliter) from 450K BeadChip and pyrosequencing analyses are shown in Figure S1.
Genome-Wide DNA Methylation in Relation to Low-Level Smoke Exposure
Using for the overall model fit and adjusting for potential covariates, we found 29,049 CpG sites to be differentially methylated in relation to a increase in cotinine among nonsmoking mothers (Excel Table S1).
Table 2 summarizes the top 20 CpG sites based on smallest adjusted p-values in relation to cotinine concentration levels among nonsmoking pregnant women in the NEST. The most significant CpG site (cg11407598) is located on chromosome 19 (, , adjusted ).
Table 2.
Top 20 CpG sites based on smallest p-value demonstrating cord blood methylation differences in cotinine concentration levels from maternal plasma samples among nonsmoking pregnant women in the newborn epigenetic study ().
CpG Site | Mapped Gene | Chr | beta | SE | Unadjusted p-value | Adjusted p-value | UCSC Referenced Gene Description |
---|---|---|---|---|---|---|---|
cg11407598 | 19 | 0.08 | |||||
cg09199225 | AGER | 6 | 1.38 | 0.16 | Advanced glycosylation end product-specific receptor | ||
cg05396900 | PRKG1 | 10 | 0.44 | 0.05 | Protein kinase, cGMP-dependent, type I | ||
cg19810457 | MAPK9 | 5 | 0.07 | Mitogen-activated protein kinase 9 | |||
cg15570148 | FHIT | 3 | 0.30 | 0.04 | Fragile histidine triad | ||
cg01588993 | 12 | 0.05 | |||||
cg01417692 | MOBKL2C | 1 | 0.01 | ||||
cg06432426 | 2 | 0.04 | |||||
cg06567155 | SLC25A15 | 13 | 0.06 | Solute carrier family 25 (mitochondrial carrier; ornithine transporter) member 15 | |||
cg06998640 | 12 | 0.01 | |||||
cg01373721 | TIMM23 | 10 | 0.13 | 0.02 | Translocase of inner mitochondrial membrane 23 homolog (yeast) | ||
cg27413290 | ZC3H3 | 8 | 0.07 | Zinc finger CCCH-type containing 3 | |||
cg14189245 | C11orf10|FEN1 | 11 | 0.46 | 0.06 | Flap structure-specific endonuclease 1 | ||
cg09719342 | STK32A | 5 | 0.12 | Serine/threonine kinase 32A | |||
cg20490386 | CTDP1 | 18 | 0.01 | CTD (carboxy-terminal domain, RNA polymerase II, polypeptide A) phosphatase, subunit 1 | |||
cg14992144 | GHRL|GHRLOS | 3 | 0.26 | 0.03 | Ghrelin opposite strand RNA conserved region|ghrelin opposite strand/antisense RNA|ghrelin/obestatin prepropeptide | ||
cg02913918 | CACNA1E | 1 | 0.02 | Calcium channel, voltage-dependent, R type, alpha 1E subunit | |||
cg07052606 | DCTPP1 | 16 | 0.27 | 0.04 | dCTP pyrophosphatase 1 | ||
cg25660646 | BSG | 19 | 0.32 | 0.04 | Basigin (Ok blood group) | ||
cg04007792 | DLGAP2 | 8 | 0.01 | Discs, large (Drosophila) homolog-associated protein 2 |
Note: Top 20 CpG sites are based on adjusted p-values, which are listed from smallest p-value to largest p-value. Beta coefficients and p-values were obtained using beta regression (see “Methods” section). Results for all FDR-significant CpGs are provided in Excel Table S1. Table 2 and Excel Table S1 show results for all FDR-significant CpGs. Model-based estimates were derived from beta regression investigating the association between methylation of umbilical cord blood from infants and secondhand smoke exposure from maternal plasma cotinine. These models were adjusted for race/ethnicity, mother’s age at delivery, maternal education, and parity, as well as technical covariates (e.g., plate, row, column), and Houseman-estimated cell proportions. The resulting p-values were corrected using Benjamini and Hochberg’s FDR method, with CpG sites with FDR considered statistically significant. Exposure contrasts for the coefficients indicate change in relation to increase in cotinine from maternal plasma. “Chr” makes reference to the chromosome number. FDR, false discovery rate; UCSC, University of California, Santa Cruz.
The second most significant CpG site (cg09199225) was associated with the AGER gene located on chromosome 6 and demonstrated the highest level of hypermethylation found in relation to a increase in plasma cotinine (, , adjusted ). In addition to this CpG site, four other CpG sites associated with AGER were identified as statistically significant. All five of these CpG sites associated with AGER demonstrated hypermethylation (Excel Table S1). The AGER gene lies within the major histocompatibility complex class III region of chromosome 6, which has been implicated in inflammatory and immune responses. It is a highly polymorphic gene that has been associated with the development of lung function, diabetes, and cardiovascular disease (Egaña-Gorroño et al. 2020; Serveaux-Dancer et al. 2019).
The third most significant CpG site (cg05396900) was also hypermethylated (, , ) with a increase in cotinine levels (Excel Table S1). This CpG site is associated with the PRKG1 gene, which encodes cyclic GMP-dependent protein kinase 1 and is involved with learning, memory, and circadian rhythm regulation (Langmesser et al. 2009). An additional five CpG sites (two showing hypermethylation and four showing hypomethylation) associated with PRKG1 showed altered methylation in relation to a increase in plasma cotinine at FDR-corrected (Excel Table S1).
When examining significant CpG probes and sorting them based on the number of sites per annotated gene, we found that there were many genes with multiple CpG sites associated with prenatal secondhand smoke exposure (Table 3). We identified 18 genes that had at least 20 statistically significant CpG sites associated with prenatal smoke exposure among nonsmoking mothers. These genes were implicated in insulin regulation (PTPRN2, which has the largest number of associated CpG sites at 87); cell signaling and regulation (MAD1L1, INPP5A, HDAC4, RASA3, RPTOR, FOXP1); neuronal processes (CAMTA1, ARHGEF10, TBCD, SHANK2); muscle, joint, organ, and/or tissue development (PRDM16, ATP11A, DIP2C, HDAC4, TNXB, SDK1); intellectual disability (CAMTA1, HDAC4, FOXP1), autism (AGAP1, SHANK2, FOXP1); tumor suppression (MAD1L1, FOXP1); and cancer (PRDM16, RPTOR, FOXP1). Relevant to the potential neurotoxic impact of secondhand smoke is the observation that the CpG site with the highest beta estimate was cg03343571 of the RNF39 gene (, , adjusted ). A total of 19 CpG sites associated with RNF39 showed significant differential methylation in relation to a increase in cotinine, with all but three CpG sites (cg26014796, cg22325294, and cg27532187) demonstrating hypomethylation. The RNF39 gene lies within the major histocompatibility complex class I region of chromosome 6 and has been implicated in multiple sclerosis (Maltby et al. 2017), schizophrenia (Carmel et al. 2020), and allergic rhinitis with and without asthma (Morin et al. 2017).
Table 3.
Genes with significant CpG sites.
Gene name | Chr | # of significant CpG Sites | Function and conditions related to changes in gene |
---|---|---|---|
PTPRN2 | 7 | 87 | Encodes a protein that is an autoantigen in type 1 diabetes (Olsson et al. 2014); regulation of insulin secretion; involvement with insulin-dependent diabetes mellitus as an autoantigen (Li et al. 1997). |
MAD1L1 | 7 | 46 | Component of mitotic spindle-assembly checkpoint that prevents onset of anaphase until all chromosomes are properly aligned at metaphase plate (Ji et al. 2018; Jin et al. 1999; Nakano et al. 2010); this may play role in cell cycle control and tumor suppression (Li et al. 2016). |
PRDM16 | 1 | 35 | Chromosomal aberration involving this gene is found in myelodysplastic syndrome and acute myeloid leukemia (Xinh et al. 2003); functions in the differentiation of brown adipose tissue (Moreno-Navarrete et al. 2018); suppressor of lung adenocarcinoma metastasis (Fei et al. 2019). |
ATP11A | 13 | 34 | Predictive marker for metachronous metastasis of colorectal cancer (Miyoshi et al. 2010); associated with bronchiectasis (persistent abnormal dilation of the bronchi) and clubbed fingers. |
DIP2C | 10 | 33 | Loss of DIP2C homolog in rat knock-outs stimulates changes in DNA methylation and epithelial-mesenchymal transition (Larsson et al. 2017); expression in breast cancer (Li et al. 2017). |
CAMTA1 | 1 | 29 | Associated with cerebellar ataxia with mental retardation (neurodevelopmental disorder characterized by mildly delayed psychomotor development, early onset of cerebellar ataxia, and intellectual disability in childhood and adulthood) (Thevenon et al. 2012); immunohistochemical marker for diagnosing epithelioid hemangioendothelioma (Shibuya et al. 2015). |
ARHGEF10 | 8 | 28 | Mutations in this gene are associated with slowed nerve conduction velocity, without any clinical signs of peripheral or central nervous system dysfunction (Verhoeven et al. 2003), and Charcot-Marie-Tooth disease (Boora et al. 2015). |
INPP5A | 10 | 28 | Mobilizes intracellular calcium and acts as a second messenger mediating cell response to various stimulation; ataxia and cerebellar degeneration in mice (Yang et al. 2015). |
TBCD | 17 | 28 | Required for correct assembly and maintenance of the mitotic spindle, and proper progression of mitosis; genetic changes can result in early-onset, progressive encephalopathy, and with brain atrophy and thin corpus callosum (an autosomal recessive disease with neurodevelopmental and neurodegenerative features) (Flex et al. 2016). |
HDAC4 | 2 | 25 | Provides instructions for making histone deacetylase 4; involved in the regulation of activity of genes involved in heart and skeletal development and nerve cell survival; mutation in the HDAC4 gene is associated with brachydactyly with no other health problems, intellectual disability, behavioral problems, and skeletal abnormalities (Williams et al. 2010). |
RASA3 | 13 | 25 | Inhibitory regulator of the Ras-cyclic AMP pathway (Molina-Ortiz et al. 2018); functions as a negative regulator of the Ras signaling pathway (Schurmans et al. 2015). |
AGAP1 | 2 | 24 | Functions as a direct regulator of the adaptor-related protein complex 3 on endosomes (Nie et al. 2005); rare variants in this gene have been identified with autism (Pacault et al. 2019). |
RPTOR | 17 | 24 | Encodes a protein that functions in cell-signaling pathway that responds to nutrient and insulin levels to regulate cell growth (Kim et al. 2002); has a positive role in maintaining cell size and mTOR protein expression, which plays a role in mRNA translation, autophagy, and cell growth; dysregulation of the mTOR pathway has been associated with cancer (Xie and Sun 2019). |
SHANK2 | 11 | 21 | May play a role in the structural and functional organization of the dendritic spine and synaptic function (MacGillavry et al. 2016) and regulate the molecular structure of Shank and the spectrum of Shank-interacting proteins in the postsynaptic densities of the adult and developing brain; mutations in this gene have been associated with autism spectrum disorder (Leblond et al. 2014). |
TNXB | 6 | 21 | Provides instructions for making tenascin-X, which plays an important role in organizing and maintaining the structure of tissues that support the body’s muscles, joints, organs, and skin/connective tissues (Mao et al. 2002); helps to regulate the production and assembly of collagen and elastic fibers in connective tissues (Kolli et al. 2019); mutations in the TNXB gene cause a very small percentage of all cases of a form of Ehlers-Danlos syndrome called the hypermobile type, which is characterized by unusually large range of joint movement (Lao et al. 2020; Micale et al. 2019); a functional variant in the TNXB promoter is associated with the risk of esophageal squamous-cell carcinoma (Yang et al. 2020). |
FOXP1 | 3 | 20 | Has an important role in the regulation of tissue- and cell type-specific gene transcription during both development and adulthood (Liu et al. 2019); may act as a tumor suppressor (Takayama et al. 2014); has been found to promote cancer stem cell-like characteristics in ovarian cancer cells (Choi et al. 2016); mutations resulting in deregulation of its expression plays an important role in the development of lung adenocarcinoma (Sheng et al. 2019) and lymphoma (Goatly et al. 2008); mutations in this gene have resulted in severe intellectual disability syndrome (Meerschaut et al. 2017; Vuillaume et al. 2018) and may play a role in speech and language disorders (Horn et al. 2010; Le Fevre et al. 2013). |
SDK1 | 7 | 20 | Codes for adhesion molecule that promotes lamina-specific synaptic connections in the retina and is expressed in specific subsets of interneurons and retinal ganglion cells (Yamagata et al. 2002); dysregulation of this protein may play an important role in podocyte dysfunction in HIV-associated nephropathy (Kaufman et al. 2004), glomerulosclerosis (Kaufman et al. 2010), and malignant mesothelioma (Cadby et al. 2013). |
Note: Chr, chromosome.
Functional enrichment analyses revealed several pathways that were enriched in genes associated with prenatal secondhand smoke exposure. Table S1 shows the top 20 pathways identified using the KEGG (Kanehisa and Goto 2000; Kanehisa et al. 2019). These pathways included neuronal signaling (e.g., cholinergic synapse, dopaminergic synapse), metabolism (e.g., gastric acid secretion, insulin secretion, aldosterone synthesis and secretion, thyroid hormone signaling), cell signaling and regulation (e.g., PI3K-Akt signaling, sphingolipid signaling, tight junction, cGMP-PKG signaling, synaptic vesicle cycle, focal adhesion, Wnt signaling), and cancer pathways [e.g., proteoglycans in cancer, pathways in cancer, extracellular matrix (ECM)-receptor interaction]. The findings highlight several pathways relevant to neurodevelopment, metabolic regulation, and cancer that are potentially affected by prenatal secondhand smoke exposure.
Epigenomic enrichment analyses indicated that there was strong enrichment within a specific class of enhancers associated with transcription start sites (TSSs), which affect protein-coding genes (Figure 1A). Notably, no other significant enrichment was observed. The low-level tobacco exposure CpGs were significantly depleted in permissive, robust, and cell type-specific enhancers (Excel Table S2 shows FANTOM Annotations from GenomeRunner) and in long noncoding RNAs and pseudogenes but were enriched in protein-coding genes (Figure 1B). Furthermore, CpGs were enriched in RNA Polymerase II binding sites (transcribing protein-coding genes) but were depleted in RNA Polymerase III (transcribing noncoding genes) (Table S2 shows Ensembl Gene Classes from GenomeRunner, and Excel Table S3 shows Transcription Factor Binding Sites from GenomeRunner). Similar to the functional enrichment pathway analysis, this analysis identified metabolic pathways, immune-related signaling, cancer pathways, and neuronal pathways among the most significant (Excel Table S4 shows KEGG Enriched Pathways). These results suggest that low-level tobacco smoke exposure may affect CpGs located near protein-coding genes and that such exposure may have a direct effect on the proteome in such a way as to influence transcription and translation of proteins.
Figure 1.
Epigenomic analysis of cotinine associated CpG sites.
Independent Validation of Associations between Cotinine Levels and DNA Methylation
As shown in Table 4, independent analysis of AGER in these participants failed to replicate the findings from the 450K BeadChip analyses (; cg09199225; , , ). However, the results for PRKG1 from the 450K BeadChip analyses were replicated for cg17079497 in the independent group of participants, with increase in cotinine significantly inversely correlated with the level of methylation at this CpG site (, , , ). As shown in Table S3, we found cg17079497 associated with PRKG1 to be significantly correlated with maternal cotinine in plasma values (, ). Quantile plots, showing associations between pyrosequencing markers (e.g., cg20720326 and cg09199225 associated with AGER and cg17079497 and cg22899538 associated with PRKG1) and maternal cotinine in plasma are shown in Figure S2.
Table 4.
Comparison of 450K and pyrosequencing analyses.
450K Beadchip analyses | Pyrosequencing analyses | ||||||||||
---|---|---|---|---|---|---|---|---|---|---|---|
Gene | CpG Site | Chr | Position | Beta | SE | FDR-corrected -value | Methylation marker | Mean proportion of methylation | Beta | SE | Regression -value |
AGER | cg09199225 | 6 | 32149260 | 1.38 | 0.16 | cg09199225 | 92.9 | 0.21 | 0.439 | ||
cg20720326 | 6 | 32152335 | 0.57 | 0.09 | cg20720326 | 87.8 | 0.14 | 0.21 | 0.490 | ||
cg26492916 | 6 | 32149401 | 0.12 | 0.03 | |||||||
cg17874413 | 6 | 32152269 | 0.08 | 0.02 | |||||||
cg27580693 | 6 | 32150024 | 0.08 | 0.03 | |||||||
PRKG1 | cg05396900 | 10 | 52750736 | 0.44 | 0.05 | ||||||
cg27380599 | 10 | 52834067 | 0.14 | 0.03 | |||||||
cg05402976 | 10 | 52993591 | 0.02 | ||||||||
cg24609819 | 10 | 52840377 | 0.06 | ||||||||
cg22899538 | 10 | 52754927 | 0.05 | cg22899538 | 90.3 | 0.47 | 0.297 | ||||
cg17079497 | 10 | 53655621 | 0.05 | cg17079497 | 84.5 | 0.45 | 0.018 |
Note: Beta coefficients and -values were obtained using beta regression for 450K Beadchip analyses (see “Methods” section). Only CpG sites that were significant for 450K Beadchip analyses at FDR are listed for AGER and PRKG1. Linear regression -values for pyrosequencing analyses are not FDR-corrected. Cotinine values under the LOD were recoded to 0 ng/mL for both 450K Beadchip analyses () and pyrosequencing analyses (). Chr, chromosome; FDR, false discovery rate; LOD, level of detection; SE, standard error.
Discussion
Similar to studies assessing active smoking during pregnancy, our results suggest that secondhand smoke exposure, measured via cotinine in maternal plasma, among a sample of nonsmoking pregnant women was significantly associated with alterations in DNA methylation of infant cord blood. In this EWAS study, we found 29,049 CpG sites to be differentially methylated in relation to a increase in cotinine among self-reported nonsmoking mothers. A number of these CpG sites were found within genes implicated in neuronal signaling, cell signaling and regulation, metabolic pathways, and cancer pathways. Further, epigenomic enrichment analyses showed a potential vulnerability to disruption in metabolic and immune-related development in response to prenatal secondhand smoke exposure. We also were able to replicate significant results from the 450K BeadChip using another independent subsample of the NEST cohort for the PRKG1 gene. Overall, results suggest that even low levels of secondhand smoke exposure during pregnancy, which likely represents exposure to other family and/or community members who smoke in the women’s environments, is related to alterations in DNA methylation of umbilical cord blood.
Methylation of CpG Sites in Relation to Low-Level Tobacco Smoke Exposure
Methylation of CpG sites associated with AHRR have been used to predict smoking status with high accuracy (Reese et al. 2017; Richmond et al. 2018; Tehranifar et al. 2018). Blood AHRR methylation at cg0557592 is inversely associated with smoking (Bojesen et al. 2017; Reynolds et al. 2015) and lung cancer (Bojesen et al. 2017), suggesting that AHRR may mediate the metabolism of xenobiotic particles like cigarette smoke. However, these studies did not examine associations in nonsmokers. In our study, which focuses on nonsmokers, we found altered methylation at 10 CpG sites associated with the AHRR gene in relation to low levels of secondhand smoke exposure. This finding suggests that DNA methylation of AHRR in infant cord blood (or from young children) may also be a useful surrogate marker of secondhand smoke exposure when direct exposure cannot be established. Of note, however, was the lack of significance for the CpG probe most often associated with smoking status, that of cg05575921 associated with AHRR (Bojesen et al. 2017; Joubert et al. 2012; Philibert et al. 2020, 2012). We did identify other CpG sites within AHRR showing significant differences in methylation in infant cord blood associated with secondhand smoke exposure during pregnancy, including one in 509 base pairs of cg05575921 (e.g., cg22103736, , , adjusted ). All of the significant probes across this region showed hypermethylation with smoke exposure (see Excel Table S1), which contrasts with the inverse correlation between methylation and smoke exposure previously reported. It is not clear why there is a directional difference in effect, but results may indicate that low-level indirect exposure elicits different effects on DNA methylation than higher-level exposure, as is the case for babies born to mothers who smoked during pregnancy. Further, the vulnerability of this region of the genome at AHRR to tobacco smoke [and to cannabis smoke exposure, see (Murphy et al. 2018)] suggests that there is inherent plasticity of the AHRR epigenome at multiple regions that may be useful for monitoring history of different levels of tobacco smoke exposure.
Six CpG sites associated with the gene PRKG1 were methylated in umbilical cord blood in relation to elevated cotinine levels in maternal plasma (Table 4). Of particular interest was cg17079497, which demonstrated statistically significant hypomethylation with both the 450K BeadChip analysis and in an independent subset of the NEST cohort by pyrosequencing. PRKG1 isoforms act as key mediators of the nitric oxide/cGMP signaling pathway and are important components of many signal transduction processes in diverse cell types. The well-studied homolog of PRKG1, foraging (for) gene in Drosophila melanogaster encodes a cGMP-dependent protein kinase (PKG). PKG activity has been found to interact with early-life stress in determining adult exploratory (i.e., the tendency to investigate a novel area) and fitness traits (i.e., fecundity) (Burns et al. 2012). Recent studies have also demonstrated genotypic variation in PRKG1 is associated with unique patterns of self-regulation in humans, such as cautious exploration of novel environments (Struk et al. 2019). Further, significant interactions between the PRKG1 C allele variant of rs1729578 and trauma exposure in relation to alcohol misuse symptoms in humans (Hawn et al. 2018; Polimanti et al. 2018), provides support for the potential role of PRKG1 in stress response-related traits in humans.
Many of the other CpGs are consistent with what has been reported in other studies examining differential methylation in relation to maternal self-reported smoking during pregnancy. Of note is one CpG site that overlapped between our study and that conducted by Joubert et al. (2016) (e.g., cg18316974 associated with GFI1). There were six FDR-significant CpGs in GFI1 associated with smoke exposure in our population. Of these CpGs, four were hypermethylated. Thus, secondhand smoke exposure was not found to be generally associated with hypermethylation in GFI1, in contrast with previous findings for sustained maternal smoking during pregnancy (de Vocht et al. 2015; Küpers et al. 2015). Results are consistent with prior studies indicating differential methylation of CpG sites associated with GFI1 between smokers and nonsmokers (Parmar et al. 2018; Philibert et al. 2013; Wan et al. 2012; Zeilinger et al. 2013). GFI1 has been found to play a role in developmental disorders; it is associated with birth weight (Küpers et al. 2015), hematopoiesis, and decreased body mass index and waist circumference (Parmar et al. 2018); and it is involved in oncogenesis (Küpers et al. 2015).
As with other environmental epigenetic studies (Reynolds et al. 2017), the effect sizes that we find in our study are small (see Figure S2). As such, the ability to detect differences in the validation cohort is limited, particularly if there was more variability in the validation cohort in the methylation levels measured across these specific CpGs. Nevertheless, small effect sizes associated with exposure are common among environmental epigenetic studies. Breton et al. (2017) posit that larger effect sizes, such as that observed in cancer, are less common because large shifts may be incompatible with continued development. The dynamic nature of the epigenome emphasizes the importance of longitudinal studies, which allow for profiling of the epigenome over both time and changing environmental exposures. Longitudinal studies will also help to improve our ability to identify small changes and determine the effect of consistent changes across time (Breton et al. 2017).
Functional Interpretation of Differentially Methylated Genes
We performed enrichment analysis to facilitate the functional interpretation of our differentially methylated genes. Pathway analysis indicated enrichment of CpG sites corresponding to genes involved in biological processes related to metabolic regulation, neuronal signaling, cell signaling and regulation, and cancer pathways. Common across these pathways is the mitogen-activated protein kinase (MAPK) signaling pathway, which plays an important role in cerebrovascular receptor plasticity (Cseh et al. 2014; Rauen 2013), as well as the regulation of gene expression, cellular growth, and survival (Knight and Irving 2014). Exposure to cigarette smoke has been shown to activate signaling pathways in airway epithelial cells, including the MAPK signaling pathway (Xu et al. 2015). Abnormal MAPK signaling may lead to increased or uncontrolled cell proliferation, resistance to apoptosis, and resistance to chemotherapy, radiotherapy, and targeted therapies through abnormal expression of pathway receptors and/or genetic mutations—all of which play an important role in the development of some cancers (Santarpia et al. 2012). This finding suggests that the MAPK pathway may be one of the mechanism by which exposure to secondhand smoke during pregnancy increases risk for cancer among offspring (Metayer et al. 2013; White et al. 2017).
Interpretation of Epigenomic Enrichment within Protein-Coding Genes Associated with Obesity-Related Traits and Obesity and Immune-Related Pathways
Similar to another study investigating enrichment analysis for the genomic features of cigarette smoking (Joehanes et al. 2016), our study demonstrated strong enrichment of CpG sites within enhancers among infants exposed to secondhand tobacco smoke in utero. Specifically, enhancers associated with transcription start sites (TSS) were affected, which supports the potential direct impact of secondhand smoke exposure on gene expression. Prior research has demonstrated that enhancers are more susceptible to methylation changes (Ziller et al. 2013) relative to other genomic regions, such as CpG islands, that are more resistant to abrupt changes in DNA methylation in response to environmental exposures (Ivanova et al. 2012). Thus, in combination with pathway analysis, our results suggest that the secondhand smoke exposure-associated CpG sites may have regulatory effects on several disease processes affecting metabolism, immunity, and cancer-related pathways.
Limitations
This prospective study should be considered in light of certain strengths and limitations. Strengths include use of a sample that includes both Black and non-Hispanic White women and their children; the use of an established, highly reproducible methylation array; and cotinine measures from maternal blood samples drawn during pregnancy to assess secondhand smoke exposure. One potential limitation of this study is related to exposure assessment. Plasma cotinine reflects a snapshot of exposure during pregnancy and may not accurately characterize exposure over the course of pregnancy or at different periods during pregnancy. Additionally, future larger studies may want to quantify varying cut points of quantitative smoke exposure to determine threshold effects on the methylation of the epigenome. Another potential limitation involves variability in DNA methylation profiles between human tissue/cell types. For this study, we used genomic DNA isolated from umbilical cord blood leukocytes. Blood samples contain interindividual variability in the number of different kinds of DNA-containing cells. Prior studies have demonstrated that disparities in methylation can arise due to the variability in cell composition when blood is used in DNA methylation analysis studies (Joubert et al. 2012; Monick et al. 2012). However, our study results show minor difference in results when adjusting for Houseman cell compositions suggesting that variability in DNA methylation due to cell composition is probably small. Another potential concern is the presence of confounding by genetic variability, which can be exacerbated in studies with small sample sizes. In addition, as is the case with many EWAS studies, the 450K BeadChip array provides measurements for CpG sites across the genome representing 99% of the RefSeq genes and 96% of CpG islands; density of CpG site coverage is still limited to the CpG sites included in the array and does not provide information on neighboring sites. Also, it is possible that the results of 450K BeadChip analyses vary between different arrays (e.g., batch effects, which were adjusted for in these analyses) or may not reflect true DNA methylation due to problematic probes and intricacies in data normalization and/or data analyses. Further, the Illumina 450K BeadChip platform may have probes that do not perform well and thus have the potential to produce erroneous results (Sugden et al. 2020). To overcome these potential limitations, next-generation sequencing can be used on CpG sites detected by the 450K BeadChip to verify changes in DNA methylation status and assess neighboring CpG sites contained within a range of specified parameters (Miyake et al. 2018). Additionally, it is possible that copy number variants (CNVs) or single nucleotide polymorphisms (SNPs) could be contributing to results. It is unlikely that SNPs are driving results, however, because CpG sites known to harbor common SNPs were excluded in analyses. It is more difficult to determine the effect of CNVs without doing a full genome assessment for copy number changes in the specimens tested, and it is unclear how common CNVs are in umbilical cord blood—suggesting that further research is needed. Finally, although our statistical models did control for race/ethnicity, age, parity, and maternal education, there is the potential for confounding by other factors (e.g., socioeconomic status, stress, diet, and alcohol consumption) that are warranted for inclusion in future follow-on studies.
Conclusion
In summary, we observed evidence of significant differences in the DNA methylation of umbilical cord blood across individual CpGs, as a function of secondhand tobacco smoke exposure measured by cotinine concentration among nonsmoking mothers during pregnancy. Our study provides preliminary evidence of effects of secondhand smoke exposure on DNA methylation in umbilical cord blood and suggests underlying molecular mechanisms that might be affected. These findings further highlight potentially harmful effects on the epigenome of babies born to nonsmoking pregnant women who are exposed to tobacco smoke from family members or in the community. Future studies are needed to replicate these findings in other study populations. Further replication would support the importance of eliminating secondhand smoke exposure during pregnancy through home and community smoke-free policies.
Supplementary Material
Acknowledgments
The Newborn Epigenetics STudy (NEST) was funded through the U.S. National Institutes of Health (R21ES014947, R01ES016772, K01CA104517, and K24DA023464) and the Duke Comprehensive Cancer Center (ACS-IRG 83-006). The Neurodevelopment and Improving Children’s Health Following Environmental Tobacco Smoke Exposure (NICHES) study was funded through the U.S. National Institutes of Health (P01ES022831) and the U.S. Environmental Protection Agency (RD-83543701). Partial support to complete the cotinine assays was provided by a grant funding through the National Cancer Institute (R01CA239595).
B.F.F. was PI for project 1 of the NICHES study and involved in the data acquisition, the study’s focus on secondhand smoke exposure and DNA methylation, design of the study, interpretation of results, and contributed to drafting and revising the manuscript. M.G.D. performed bioinformatics/statistical analyses of methylation data, and generated results tables and figures. E.K.D. contributed to the statistical analysis of pyrosequencing data, as well as conducting background literature for the introduction and the development of the results, methods, and discussion. C.G. and Z.H. facilitated sample preparation for 450K BeadChip data generation and generated the pyrosequencing data. S.K.M. was involved in study conception and design, data interpretation, and contributed to drafting and revising the manuscript. J.Z. supervised cotinine assays and reviewed the manuscript. C.H. was the PI and was involved with the design of the NEST cohort. S.H.K. was involved in the data acquisition of the NICHES study. All of the authors discussed the results, contributed to the discussion, and helped in the preparing and editing of this manuscript.
Study data is available from the corresponding author (B.F.F.) upon reasonable request.
References
- Benjamini Y, Hochberg Y. 1995. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc Series B (Methodological) 57(1):289–300, 10.1111/j.2517-6161.1995.tb02031.x. [DOI] [Google Scholar]
- Benowitz NL, Bernert JT, Caraballo RS, Holiday DB, Wang J. 2009. Optimal serum cotinine levels for distinguishing cigarette smokers and nonsmokers within different racial/ethnic groups in the United States between 1999 and 2004. Am J Epidemiol 169(2):236–248, PMID: 19019851, 10.1093/aje/kwn301. [DOI] [PubMed] [Google Scholar]
- Bergens MA, Pittman GS, Thompson IJB, Campbell MR, Wang X, Hoyo C, et al. 2019. Smoking-associated AHRR demethylation in cord blood DNA: impact of CD235a+ nucleated red blood cells. Clin Epigenetics 11(1):87, PMID: 31182156, 10.1186/s13148-019-0686-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bernert JT, Jacob P, Holiday DB, Benowitz NL, Sosnoff CS, Doig MV, et al. 2009. Interlaboratory comparability of serum cotinine measurements at smoker and nonsmoker concentration levels: a round-robin study. Nicotine Tob Res 11(12):1458–1466, PMID: 19933777, 10.1093/ntr/ntp161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bibikova M, Barnes B, Tsan C, Ho V, Klotzle B, Le JM, et al. 2011. High density DNA methylation array with single CpG site resolution. Genomics 98(4):288–295, PMID: 21839163, 10.1016/j.ygeno.2011.07.007. [DOI] [PubMed] [Google Scholar]
- Bojesen SE, Timpson N, Relton C, Davey Smith G, Nordestgaard BG. 2017. AHRR (cg05575921) hypomethylation marks smoking behaviour, morbidity and mortality. Thorax 72(7):646–653, PMID: 28100713, 10.1136/thoraxjnl-2016-208789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boora GK, Kulkarni AA, Kanwar R, Beyerlein P, Qin R, Banck MS, et al. 2015. Association of the Charcot-Marie-Tooth disease gene ARHGEF10 with paclitaxel induced peripheral neuropathy in NCCTG N08CA (alliance). J Neurol Sci 357(1–2):35–40, PMID: 26143528, 10.1016/j.jns.2015.06.056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Breitling LP, Yang R, Korn B, Burwinkel B, Brenner H. 2011. Tobacco-smoking-related differential DNA methylation: 27K discovery and replication. Am J Hum Genet 88(4):450–457, PMID: 21457905, 10.1016/j.ajhg.2011.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Breton CV, Byun H-M, Wenten M, Pan F, Yang A, Gilliland FD. 2009. Prenatal tobacco smoke exposure affects global and gene-specific DNA methylation. Am J Respir Crit Care Med 180(5):462–467, PMID: 19498054, 10.1164/rccm.200901-0135OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Breton CV, Marsit CJ, Faustman E, Nadeau K, Goodrich JM, Dolinoy DC, et al. 2017. Small-magnitude effect sizes in epigenetic end points are important in children’s environmental health studies: the Children’s Environmental Health and Disease Prevention Research Center’s Epigenetics Working Group. Environ Health Perspect 125(4):511–526, PMID: 28362264, 10.1289/EHP595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Burns JG, Svetec N, Rowe L, Mery F, Dolan MJ, Thom W, et al. 2012. Gene-environment interplay in Drosophila melanogaster: Chronic food deprivation in early life affects adult exploratory and fitness traits. Proc Natl Acad Sci USA 109:17239–17244, 10.1073/pnas.1121265109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cadby G, Mukherjee S, Musk AWB, Reid A, Garlepp M, Dick I, et al. 2013. A genome-wide association study for malignant mesothelioma risk. Lung Cancer 82(1):1–8, PMID: 23827383, 10.1016/j.lungcan.2013.04.018. [DOI] [PubMed] [Google Scholar]
- Carmel M, Michaelovsky E, Weinberger R, Frisch A, Mekori-Domachevsky E, Gothelf D, et al. 2020. Differential methylation of imprinting genes and MHC locus in 22q11.2 deletion syndrome-related schizophrenia spectrum disorders. World J Biol Psychiatry 1–12, PMID: 32212948, 10.1080/15622975.2020.1747113. [DOI] [PubMed] [Google Scholar]
- Centers for Disease Control and Prevention. 2020. Substance Use During Pregnancy CDC. https://www.cdc.gov/reproductivehealth/maternalinfanthealth/substance-abuse/substance-abuse-during-pregnancy.htm [accessed 2 February 2021].
- Chen EY, Tan CM, Kou Y, Duan Q, Wang Z, Meirelles GV, et al. 2013a. Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool. BMC Bioinformatics 14:128, PMID: 23586463, 10.1186/1471-2105-14-128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen Y, Lemire M, Choufani S, Butcher DT, Grafodatskaya D, Zanke BW, et al. 2013b. Discovery of cross-reactive probes and polymorphic CpGs in the Illumina Infinium HumanMethylation450 microarray. Epigenetics 8(2):203–209, PMID: 23314698, 10.4161/epi.23470. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Choi EJ, Seo EJ, Kim DK, Lee SI, Kwon YW, Jang IH, et al. 2016. FOXP1 functions as an oncogene in promoting cancer stem cell-like characteristics in ovarian cancer cells. Oncotarget 7(3):3506–3519, PMID: 26654944, 10.18632/oncotarget.6510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cribari-Neto F, Zeileis A. 2010. Beta regression in R. J Stat Soft 34(2):1–24, 10.18637/jss.v034.i02. [DOI] [Google Scholar]
- Cseh B, Doma E, Baccarini M. 2014. “RAF” neighborhood: protein–protein interaction in the Raf/Mek/Erk pathway. FEBS Letters 588(15):2398–2406, PMID: 24937142, 10.1016/j.febslet.2014.06.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Davis S, Milke S, Tiche T, Bootwalla M. 2020. methylumi: Handle Illumina methylation data version 2.36.0 from Bioconductor. https://bioconductor.org/packages/release/bioc/html/methylumi.html.
- de Vocht F, Simpkin AJ, Richmond RC, Relton C, Tilling K. 2015. Assessment of offspring DNA methylation across the lifecourse associated with prenatal maternal smoking using Bayesian mixture modelling. Int J Environ Res Public Health 12(11):14461–14476, PMID: 26580635, 10.3390/ijerph121114461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dempsey DA, Meyers MJ, Oh SS, Nguyen EA, Fuentes-Afflick E, Wu AHB, et al. 2012. Determination of tobacco smoke exposure by plasma cotinine levels in infants and children attending urban public hospital clinics. Arch Pediatr Adolesc Med 166(9):851–856, PMID: 22566513, 10.1001/archpediatrics.2012.170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Do EK, Green TL, Prom-Wormley EC, Fuemmeler BF. 2018. Social determinants of smoke exposure during pregnancy: findings from waves 1 & 2 of the population assessment of tobacco and health (PATH) study. Prev Med Rep 12:312–320, PMID: 30406010, 10.1016/j.pmedr.2018.10.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dogan MV, Shields B, Cutrona C, Gao L, Gibbons FX, Simons R, et al. 2014. The effect of smoking on DNA methylation of peripheral blood mononuclear cells from African American women. BMC Genomics 15:151, PMID: 24559495, 10.1186/1471-2164-15-151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dozmorov MG, Cara LR, Giles CB, Wren JD. 2012. GenomeRunner: automating genome exploration. Bioinformatics 28(3):419–420, PMID: 22155868, 10.1093/bioinformatics/btr666. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dozmorov MG, Cara LR, Giles CB, Wren JD. 2016. GenomeRunner web server: regulatory similarity and differences define the functional impact of SNP sets. Bioinformatics 32(15):2256–2263, PMID: 27153607, 10.1093/bioinformatics/btw169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Egaña-Gorroño L, López-Díez R, Yepuri G, Ramirez LS, Reverdatto S, Gugger PF, et al. 2020. Receptor for advanced glycation end products (RAGE) and mechanisms and therapeutic opportunities in diabetes and cardiovascular disease: insights from human subjects and animal models. Front Cardiovasc Med 7:37, PMID: 32211423, 10.3389/fcvm.2020.00037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Elliott HR, Tillin T, McArdle WL, Ho K, Duggirala A, Frayling TM, et al. 2014. Differences in smoking associated DNA methylation patterns in South Asians and Europeans. Clin Epigenetics 6(1):4, PMID: 24485148, 10.1186/1868-7083-6-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fei L-R, Huang W-J, Wang Y, Lei L, Li Z-H, Zheng Y-W, et al. 2019. PRDM16 functions as a suppressor of lung adenocarcinoma metastasis. J Exp Clin Cancer Res 38(1):35, PMID: 30683132, 10.1186/s13046-019-1042-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Flex E, Niceta M, Cecchetti S, Thiffault I, Au MG, Capuano A, et al. 2016. Biallelic mutations in TBCD, encoding the tubulin folding cofactor D, perturb microtubule dynamics and cause early-onset encephalopathy. Am J Hum Genet 99(4):962–973, PMID: 27666370, 10.1016/j.ajhg.2016.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Garg M, Garrison L, Leeman L, Hamidovic A, Borrego M, Rayburn WF, et al. 2016. Validity of self-reported drug use information among pregnant women. Matern Child Health J 20(1):41–47, PMID: 26175273, 10.1007/s10995-015-1799-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goatly A, Bacon CM, Nakamura S, Ye H, Kim I, Brown PJ, et al. 2008. FOXP1 abnormalities in lymphoma: translocation breakpoint mapping reveals insights into deregulated transcriptional control. Mod Pathol 21(7):902–911, PMID: 18487996, 10.1038/modpathol.2008.74. [DOI] [PubMed] [Google Scholar]
- Gonzalez-Nahm S, Mendez MA, Benjamin-Neelon SE, Murphy SK, Hogan VK, Rowley DL, et al. 2018. DNA methylation of imprinted genes at birth is associated with child weight status at birth, 1 year, and 3 years. Clinical Epigenetics 10:90, PMID: 29988473, 10.1186/s13148-018-0521-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hawn SE, Sheerin CM, Webb BT, Peterson RE, Do EK, Dick D, et al. 2018. Replication of the interaction of PRKG1 and trauma exposure on alcohol misuse in an independent African American sample. J Trauma Stress 31(6):927–932, PMID: 30376604, 10.1002/jts.22339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Horn D, Kapeller J, Rivera-Brugués N, Moog U, Lorenz-Depiereux B, Eck S, et al. 2010. Identification of FOXP1 deletions in three unrelated patients with mental retardation and significant speech and language deficits. Hum Mutat 31(11):E1851–E1860, PMID: 20848658, 10.1002/humu.21362. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Houseman EA, Accomando WP, Koestler DC, Christensen BC, Marsit CJ, Nelson HH, et al. 2012. DNA methylation arrays as surrogate measures of cell mixture distribution. BMC Bioinformatics 13:86, PMID: 22568884, 10.1186/1471-2105-13-86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hoyo C, Murtha AP, Schildkraut JM, Jirtle R, Demark-Wahnefried W, Forman MR, et al. 2011. Methylation variation at IGF2 differentially methylated regions and maternal folic acid use before and during pregnancy. Epigenetics 6(7):928–936, PMID: 21636975, 10.4161/epi.6.7.16263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ivanova E, Chen J-H, Segonds-Pichon A, Ozanne SE, Kelsey G. 2012. DNA methylation at differentially methylated regions of imprinted genes is resistant to developmental programming by maternal nutrition. Epigenetics 7(10):1200–1210, PMID: 22968513, 10.4161/epi.22141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ivorra C, Fraga MF, Bayón GF, Fernández AF, Garcia-Vicent C, Chaves FJ, et al. 2015. DNA methylation patterns in newborns exposed to tobacco in utero. J Transl Med 13(1):25, PMID: 25623364, 10.1186/s12967-015-0384-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jacob P, Yu L, Duan M, Ramos L, Yturralde O, Benowitz NL. 2011. Determination of the nicotine metabolites cotinine and trans-3′-hydroxycotinine in biologic fluids of smokers and non-smokers using liquid chromatography–tandem mass spectrometry: biomarkers for tobacco smoke exposure and for phenotyping cytochrome P450 2A6 activity. J Chromatogr B Analyt Technol Biomed Life Sci 879(3–4):267–276, PMID: 21208832, 10.1016/j.jchromb.2010.12.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ji W, Luo Y, Ahmad E, Liu S-T. 2018. Direct interactions of mitotic arrest deficient 1 (MAD1) domains with each other and MAD2 conformers are required for mitotic checkpoint signaling. J Biol Chem 293(2):484–496, PMID: 29162720, 10.1074/jbc.RA117.000555. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jin DY, Kozak CA, Pangilinan F, Spencer F, Green ED, Jeang KT. 1999. Mitotic checkpoint locus MAD1L1 maps to human chromosome 7p22 and mouse chromosome 5. Genomics 55(3):363–364, PMID: 10049595, 10.1006/geno.1998.5654. [DOI] [PubMed] [Google Scholar]
- Joehanes R, Just AC, Marioni RE, Pilling LC, Reynolds LM, Mandaviya PR, et al. 2016. Epigenetic signatures of cigarette smoking. Circ Cardiovasc Genet 9(5):436–447, PMID: 27651444, 10.1161/CIRCGENETICS.116.001506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Joubert BR, Felix JF, Yousefi P, Bakulski KM, Just AC, Breton C, et al. 2016. DNA methylation in newborns and maternal smoking in pregnancy: genome-wide consortium Meta-analysis. Am J Hum Genet 98(4):680–696, PMID: 27040690, 10.1016/j.ajhg.2016.02.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Joubert BR, Håberg SE, Nilsen RM, Wang X, Vollset SE, Murphy SK, et al. 2012. 450K epigenome-wide scan identifies differential DNA methylation in newborns related to maternal smoking during pregnancy. Environ Health Perspect 120(10):1425–1431, PMID: 22851337, 10.1289/ehp.1205412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kanehisa M, Goto S. 2000. KEGG: Kyoto Encyclopedia of Genes and Genomes. Nucleic Acids Res 28(1):27–30, PMID: 10592173, 10.1093/nar/28.1.27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kanehisa M, Sato Y, Furumichi M, Morishima K, Tanabe M. 2019. New approach for understanding genome variations in KEGG. Nucleic Acids Res 47(D1):D590–D595, PMID: 30321428, 10.1093/nar/gky962. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kaufman L, Hayashi K, Ross MJ, Ross MD, Klotman PE. 2004. Sidekick-1 is upregulated in glomeruli in HIV-associated nephropathy. J Am Soc Nephrol 15(7):1721–1730, PMID: 15213259, 10.1097/01.ASN.0000128975.28958.C2. [DOI] [PubMed] [Google Scholar]
- Kaufman L, Potla U, Coleman S, Dikiy S, Hata Y, Kurihara H, et al. 2010. Up-regulation of the homophilic adhesion molecule sidekick-1 in podocytes contributes to glomerulosclerosis. J Biol Chem 285(33):25677–25685, PMID: 20562105, 10.1074/jbc.M110.133959. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim D-H, Sarbassov DD, Ali SM, King JE, Latek RR, Erdjument-Bromage H, et al. 2002. mTOR interacts with raptor to form a nutrient-sensitive complex that signals to the cell growth machinery. Cell 110(2):163–175, PMID: 12150925, 10.1016/S0092-8674(02)00808-5. [DOI] [PubMed] [Google Scholar]
- Kirchner TR, Cantrell J, Anesetti-Rothermel A, Ganz O, Vallone DM, Abrams DB. 2013. Geospatial exposure to point-of-sale tobacco. Am J Prev Med 45(4):379–385, PMID: 24050412, 10.1016/j.amepre.2013.05.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Knight T, Irving JAE. 2014. Ras/Raf/MEK/ERK pathway activation in childhood acute lymphoblastic leukemia and its therapeutic targeting. Front Oncol 4:160, PMID: 25009801, 10.3389/fonc.2014.00160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kolli V, Kim H, Rao H, Lao Q, Gaynor A, Milner JD, et al. 2019. Measurement of serum tenascin-X in patients with congenital adrenal hyperplasia at risk for Ehlers-Danlos contiguous gene deletion syndrome CAH-X. BMC Res Notes 12(1):711, PMID: 31666125, 10.1186/s13104-019-4753-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Küpers LK, Xu X, Jankipersadsing SA, Vaez A, la Bastide-van Gemert S, Scholtens S, et al. 2015. DNA methylation mediates the effect of maternal smoking during pregnancy on birthweight of the offspring. Int J Epidemiol 44(4):1224–1237, PMID: 25862628, 10.1093/ije/dyv048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Langmesser S, Franken P, Feil S, Emmenegger Y, Albrecht U, Feil R. 2009. cGMP-dependent protein kinase type I is implicated in the regulation of the timing and quality of sleep and wakefulness. PLoS One 4(1):e4238, PMID: 19156199, 10.1371/journal.pone.0004238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lao Q, Mallappa A, Rueda FF, Joyal E, Veeraraghavan P, Chen W, et al. 2020. A TNXB splice donor site variant as a cause of hypermobility type Ehlers-Danlos syndrome in patients with congenital adrenal hyperplasia. Mol Genet Genomic Med 9(2):e1556, PMID: 33332743, 10.1002/mgg3.1556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Larsson C, Ali MA, Pandzic T, Lindroth AM, He L, Sjöblom T. 2017. Loss of DIP2C in RKO cells stimulates changes in DNA methylation and epithelial-mesenchymal transition. BMC Cancer 17(1):487, PMID: 28716088, 10.1186/s12885-017-3472-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Le Fevre AK, Taylor S, Malek NH, Horn D, Carr CW, Abdul-Rahman OA, et al. 2013. FOXP1 mutations cause intellectual disability and a recognizable phenotype. Am J Med Genet A 161A (12):3166–3175, PMID: 24214399, 10.1002/ajmg.a.36174. [DOI] [PubMed] [Google Scholar]
- Leblond CS, Nava C, Polge A, Gauthier J, Huguet G, Lumbroso S, et al. 2014. Meta-analysis of SHANK mutations in autism spectrum disorders: a gradient of severity in cognitive impairments. PLoS Genet 10(9):e1004580, PMID: 25188300, 10.1371/journal.pgen.1004580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee KWK, Richmond R, Hu P, French L, Shin J, Bourdon C, et al. 2015. Prenatal exposure to maternal cigarette smoking and DNA methylation: epigenome-wide association in a discovery sample of adolescents and replication in an independent cohort at birth through 17 years of age. Environ Health Perspect 123(2):193–199, PMID: 25325234, 10.1289/ehp.1408614. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li Q, Borovitskaya AE, DeSilva MG, Wasserfall C, Maclaren NK, Notkins AL, et al. 1997. Autoantigens in insulin-dependent diabetes mellitus: molecular cloning and characterization of human IA-2 beta. Proc Assoc Am Physicians 109(4):429–439, PMID: 9220540. [PubMed] [Google Scholar]
- Li D, Meng Q, Zhang H, Feng T, Liu M, Cai L. 2016. Mitotic arrest deficient-like 1 is correlated with poor prognosis in small-cell lung cancer after surgical resection. Tumor Biol 37(4):4393–4398, PMID: 26499943, 10.1007/s13277-015-4302-5. [DOI] [PubMed] [Google Scholar]
- Li J, Ping JL, Ma B, Chen YR, Li LQ. 2017. DIP2C expression in breast cancer and its clinical significance. Pathol Res Pract 213(11):1394–1399, PMID: 28964575, 10.1016/j.prp.2017.09.007. [DOI] [PubMed] [Google Scholar]
- Liu P, Huang S, Ling S, Xu S, Wang F, Zhang W, et al. 2019. Foxp1 controls brown/beige adipocyte differentiation and thermogenesis through regulating β3-AR desensitization. Nat Commun 10(1):5070, PMID: 31699980, 10.1038/s41467-019-12988-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu Y, Murphy SK, Murtha AP, Fuemmeler BF, Schildkraut J, Huang Z, et al. 2012. Depression in pregnancy, infant birth weight and DNA methylation of imprint regulatory elements. Epigenetics 7(7):735–746, PMID: 22677950, 10.4161/epi.20734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- MacGillavry HD, Kerr JM, Kassner J, Frost NA, Blanpied TA. 2016. Shank-cortactin interactions control actin dynamics to maintain flexibility of neuronal spines and synapses. Eur J Neurosci 43(2):179–193, PMID: 26547831, 10.1111/ejn.13129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maltby VE, Lea RA, Sanders KA, White N, Benton MC, Scott RJ, et al. 2017. Differential methylation at MHC in CD4+ T cells is associated with multiple sclerosis independently of HLA-DRB1. Clin Epigenet 9(1):71, PMID: 28729889, 10.1186/s13148-017-0371-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mao JR, Taylor G, Dean WB, Wagner DR, Afzal V, Lotz JC, et al. 2002. Tenascin-X deficiency mimics Ehlers-Danlos syndrome in mice through alteration of collagen deposition. Nat Genet 30(4):421–425, PMID: 11925569, 10.1038/ng850. [DOI] [PubMed] [Google Scholar]
- Meerschaut I, Rochefort D, Revençu N, Pètre J, Corsello C, Rouleau GA, et al. 2017. FOXP1-related intellectual disability syndrome: a recognisable entity. J Med Genet 54(9):613–623, PMID: 28735298, 10.1136/jmedgenet-2017-104579. [DOI] [PubMed] [Google Scholar]
- Metayer C, Zhang L, Wiemels JL, Bartley K, Schiffman J, Ma X, et al. 2013. Tobacco smoke exposure and the risk of childhood acute lymphoblastic and myeloid leukemias by cytogenetic subtype. Cancer Epidemiol Biomarkers Prev 22(9):1600–1611, PMID: 23853208, 10.1158/1055-9965.EPI-13-0350. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Micale L, Guarnieri V, Augello B, Palumbo O, Agolini E, Sofia VM, et al. 2019. Novel TNXB variants in two Italian patients with classical-like Ehlers-Danlos syndrome. Genes (Basel) 10(12):967, PMID: 31775249, 10.3390/genes10120967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miyake K, Kawaguchi A, Miura R, Kobayashi S, Tran NQV, Kobayashi S, et al. 2018. Association between DNA methylation in cord blood and maternal smoking: the Hokkaido Study on Environment and Children’s Health. Sci Rep 8(1):5654, PMID: 29618728, 10.1038/s41598-018-23772-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miyoshi N, Ishii H, Mimori K, Tanaka F, Nagai K, Uemura M, et al. 2010. ATP11A is a novel predictive marker for metachronous metastasis of colorectal cancer. Oncol Rep 23(2):505–510, PMID: 20043114, 10.3892/or_00000662. [DOI] [PubMed] [Google Scholar]
- Molina-Ortiz P, Orban T, Martin M, Habets A, Dequiedt F, Schurmans S. 2018. Rasa3 controls turnover of endothelial cell adhesion and vascular lumen integrity by a Rap1-dependent mechanism. PLoS Genet 14(1):e1007195, PMID: 29381707, 10.1371/journal.pgen.1007195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Monick MM, Beach SRH, Plume J, Sears R, Gerrard M, Brody GH, et al. 2012. Coordinated changes in AHRR methylation in lymphoblasts and pulmonary macrophages from smokers. Am J Med Genet B Neuropsychiatr Genet 159B(2):141–151, PMID: 22232023, 10.1002/ajmg.b.32021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moreno-Navarrete JM, Serino M, Blasco-Baque V, Azalbert V, Barton RH, Cardellini M, et al. 2018. Gut microbiota interacts with markers of adipose tissue browning, insulin action and plasma acetate in morbid obesity. Mol Nutr Food Res 62(3):1700721, PMID: 29105287, 10.1002/mnfr.201700721. [DOI] [PubMed] [Google Scholar]
- Morin A, Laviolette M, Pastinen T, Boulet L-P, Laprise C. 2017. Combining omics data to identify genes associated with allergic rhinitis. Clin Epigenet 9(1):3, PMID: 28149331, 10.1186/s13148-017-0310-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Murphy SK, Huang Z, Hoyo C. 2012. Differentially methylated regions of imprinted genes in prenatal, perinatal and postnatal human tissues. PLoS One 7(7):e40924, PMID: 22808284, 10.1371/journal.pone.0040924. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Murphy SK, Itchon-Ramos N, Visco Z, Huang Z, Grenier C, Schrott R, et al. 2018. Cannabinoid exposure and altered DNA methylation in rat and human sperm. Epigenetics 13(12):1208–1221, PMID: 30521419, 10.1080/15592294.2018.1554521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nakano H, Funasaka T, Hashizume C, Wong RW. 2010. Nucleoporin translocated promoter region (Tpr) associates with dynein complex, preventing chromosome lagging formation during mitosis. J Biol Chem 285(14):10841–10849, PMID: 20133940, 10.1074/jbc.M110.105890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nie Z, Fei J, Premont RT, Randazzo PA. 2005. The Arf GAPs AGAP1 and AGAP2 distinguish between the adaptor protein complexes AP-1 and AP-3. J Cell Sci 118(pt 15):3555–3566, PMID: 16079295, 10.1242/jcs.02486. [DOI] [PubMed] [Google Scholar]
- Noël A, Xiao R, Perveen Z, Zaman H, Le Donne V, Penn A. 2017. Sex-specific lung functional changes in adult mice exposed only to second-hand smoke in utero. Respir Res 18(1):104, PMID: 28651580, 10.1186/s12931-017-0591-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Olsson AH, Volkov P, Bacos K, Dayeh T, Hall E, Nilsson EA, et al. 2014. Genome-wide associations between genetic and epigenetic variation influence mRNA expression and insulin secretion in human pancreatic islets. PLoS Genet 10(11):e1004735, PMID: 25375650, 10.1371/journal.pgen.1004735. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pacault M, Nizon M, Pichon O, Vincent M, Le Caignec C, Isidor B. 2019. A de novo 2q37.2 deletion encompassing AGAP1 and SH3BP4 in a patient with autism and intellectual disability. Eur J Med Genet 62(12):103586, PMID: 30472483, 10.1016/j.ejmg.2018.11.020. [DOI] [PubMed] [Google Scholar]
- Parmar P, Lowry E, Cugliari G, Suderman M, Wilson R, Karhunen V, et al. 2018. Association of maternal prenatal smoking GFI1-locus and cardio-metabolic phenotypes in 18,212 adults. EBioMedicine 38:206–216, PMID: 30442561, 10.1016/j.ebiom.2018.10.066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Philibert RA, Beach SRH, Brody GH. 2012. Demethylation of the aryl hydrocarbon receptor repressor as a biomarker for nascent smokers. Epigenetics 7(11):1331–1338, PMID: 23070629, 10.4161/epi.22520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Philibert RA, Beach SRH, Lei M-K, Brody GH. 2013. Changes in DNA methylation at the aryl hydrocarbon receptor repressor may be a new biomarker for smoking. Clin Epigenet 5(1):19, PMID: 24120260, 10.1186/1868-7083-5-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Philibert R, Dogan M, Beach SRH, Mills JA, Long JD. 2020. AHRR methylation predicts smoking status and smoking intensity in both saliva and blood DNA. Am J Med Genet B Neuropsychiatr Genet 183(1):51–60, PMID: 31456352, 10.1002/ajmg.b.32760. [DOI] [PubMed] [Google Scholar]
- Pidsley R, Zotenko E, Peters TJ, Lawrence MG, Risbridger GP, Molloy P, et al. 2016. Critical evaluation of the Illumina MethylationEPIC BeadChip microarray for whole-genome DNA methylation profiling. Genome Biol 17(1):208, PMID: 27717381, 10.1186/s13059-016-1066-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Polimanti R, Kaufman J, Zhao H, Kranzler HR, Ursano RJ, Kessler RC, et al. 2018. A genome-wide gene-by-trauma interaction study of alcohol misuse in two independent cohorts identifies PRKG1 as a risk locus. Mol Psychiatry 23(1):154–160, PMID: 28265120, 10.1038/mp.2017.24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Prince C, Hammerton G, Taylor AE, Anderson EL, Timpson NJ, Davey Smith G, et al. 2019. Investigating the impact of cigarette smoking behaviours on DNA methylation patterns in adolescence. Hum Mol Genet 28(1):155–165, PMID: 30215712, 10.1093/hmg/ddy316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rauen KA. 2013. The RASopathies. Annu Rev Genomics Hum Genet 14:355–369, PMID: 23875798, 10.1146/annurev-genom-091212-153523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reese SE, Zhao S, Wu MC, Joubert BR, Parr CL, Håberg SE, et al. 2017. DNA methylation score as a biomarker in newborns for sustained maternal smoking during pregnancy. Environ Health Perspect 125(4):760–766, PMID: 27323799, 10.1289/EHP333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reinius LE, Acevedo N, Joerink M, Pershagen G, Dahlén S-E, Greco D, et al. 2012. Differential DNA methylation in purified human blood cells: implications for cell lineage and studies on disease susceptibility. PLoS One 7(7):e41361, PMID: 22848472, 10.1371/journal.pone.0041361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reynolds LM, Magid HS, Chi GC, Lohman K, Barr RG, Kaufman JD, et al. 2017. Secondhand tobacco smoke exposure associations with DNA methylation of the aryl hydrocarbon receptor repressor. Nicotine Tob Res 19:442–451, PMID: 27613907, 10.1093/ntr/ntw219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reynolds LM, Wan M, Ding J, Taylor JR, Lohman K, Su D, et al. 2015. DNA methylation of the aryl hydrocarbon receptor repressor associations with cigarette smoking and subclinical atherosclerosis. Circ Cardiovasc Genet 8(5):707–716, PMID: 26307030, 10.1161/CIRCGENETICS.115.001097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Richmond RC, Simpkin AJ, Woodward G, Gaunt TR, Lyttleton O, McArdle WL, et al. 2015. Prenatal exposure to maternal smoking and offspring DNA methylation across the lifecourse: findings from the Avon Longitudinal Study of Parents and Children (ALSPAC). Hum Mol Genet 24(8):2201–2217, PMID: 25552657, 10.1093/hmg/ddu739. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Richmond RC, Suderman M, Langdon R, Relton CL, Davey Smith G. 2018. DNA methylation as a marker for prenatal smoke exposure in adults. Int J Epidemiol 47(4):1120–1130, PMID: 29860346, 10.1093/ije/dyy091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Santarpia L, Lippman SM, El-Naggar AK. 2012. Targeting the MAPK-RAS-RAF signaling pathway in cancer therapy. Expert Opin Ther Targets 16(1):103–119, PMID: 22239440, 10.1517/14728222.2011.645805. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schechter JC, Fuemmeler BF, Hoyo C, Murphy SK, Zhang JJ, Kollins SH. 2018. Impact of smoking ban on passive smoke exposure in pregnant non-smokers in the southeastern United States. Int J Environ Res Public Health 15(1):83, PMID: 29316617, 10.3390/ijerph15010083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schurmans S, Polizzi S, Scoumanne A, Sayyed S, Molina-Ortiz P. 2015. The Ras/Rap GTPase activating protein RASA3: from gene structure to in vivo functions. Adv Biol Regul 57:153–161, PMID: 25294679, 10.1016/j.jbior.2014.09.006. [DOI] [PubMed] [Google Scholar]
- Serveaux-Dancer M, Jabaudon M, Creveaux I, Belville C, Blondonnet R, Gross C, et al. 2019. Pathological implications of receptor for advanced glycation end-product (AGER) gene polymorphism. Dis Markers 2019:2067353, PMID: 30863465, 10.1155/2019/2067353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sheng H, Li X, Xu Y. 2019. Knockdown of FOXP1 promotes the development of lung adenocarcinoma. Cancer Biol Ther 20(4):537–545, PMID: 30409062, 10.1080/15384047.2018.1537999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shenker NS, Polidoro S, van Veldhoven K, Sacerdote C, Ricceri F, Birrell MA, et al. 2013. Epigenome-wide association study in the European Prospective Investigation into Cancer and Nutrition (EPIC-Turin) identifies novel genetic loci associated with smoking. Hum Mol Genet 22(5):843–851, PMID: 23175441, 10.1093/hmg/dds488. [DOI] [PubMed] [Google Scholar]
- Shibuya R, Matsuyama A, Shiba E, Harada H, Yabuki K, Hisaoka M. 2015. CAMTA1 is a useful immunohistochemical marker for diagnosing epithelioid haemangioendothelioma. Histopathology 67(6):827–835, PMID: 25879300, 10.1111/his.12713. [DOI] [PubMed] [Google Scholar]
- Struk AA, Mugon J, Huston A, Scholer AA, Stadler G, Higgins ET, et al. 2019. Self-regulation and the foraging gene (PRKG1) in humans. Proc Natl Acad Sci USA 116(10):4434–4439, PMID: 30782798, 10.1073/pnas.1809924116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sugden K, Hannon EJ, Arseneault L, Belsky DW, Corcoran DL, Fisher HL, et al. 2020. Patterns of reliability: assessing the reproducibility and integrity of DNA methylation measurement. Patterns (NY) 1(2):100014, PMID: 33205092, 10.1016/j.patter.2020.100014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Suter MA, Aagaard K. 2012. What changes in DNA methylation take place in individuals exposed to maternal smoking in utero? Epigenomics 4(2):115–118, PMID: 22449181, 10.2217/epi.12.7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Suter MA, Anders AM, Aagaard KM. 2013. Maternal smoking as a model for environmental epigenetic changes affecting birthweight and fetal programming. Mol Hum Reprod 19(1):1–6, PMID: 23139402, 10.1093/molehr/gas050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Takayama K-I, Suzuki T, Tsutsumi S, Fujimura T, Takahashi S, Homma Y, et al. 2014. Integrative analysis of FOXP1 function reveals a tumor-suppressive effect in prostate cancer. Mol Endocrinol 28(12):2012–2024, PMID: 25329375, 10.1210/me.2014-1171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tehranifar P, Wu H-C, McDonald JA, Jasmine F, Santella RM, Gurvich I, et al. 2018. Maternal cigarette smoking during pregnancy and offspring DNA methylation in midlife. Epigenetics 13(2):129–134, PMID: 28494218, 10.1080/15592294.2017.1325065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thevenon J, Lopez E, Keren B, Heron D, Mignot C, Altuzarra C, et al. 2012. Intragenic CAMTA1 rearrangements cause non-progressive congenital ataxia with or without intellectual disability. J Med Genet 49(6):400–408, PMID: 22693284, 10.1136/jmedgenet-2012-100856. [DOI] [PubMed] [Google Scholar]
- Triche TJ, Laird PW, Siegmund KD. 2016. Beta regression improves the detection of differential DNA methylation for epigenetic epidemiology. bioRxiv, 10.1101/054643. [DOI] [Google Scholar]
- Verhoeven K, De Jonghe P, Van de Putte T, Nelis E, Zwijsen A, Verpoorten N, et al. 2003. Slowed conduction and thin myelination of peripheral nerves associated with mutant rho Guanine-nucleotide exchange factor 10. Am J Hum Genet 73(4):926–932, PMID: 14508709, 10.1086/378159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vuillaume M-L, Cogné B, Jeanne M, Boland A, Ung D-C, Quinquis D, et al. 2018. Whole genome sequencing identifies a de novo 2.1 Mb balanced paracentric inversion disrupting FOXP1 and leading to severe intellectual disability. Clin Chim Acta 485:218–223, PMID: 29969624, 10.1016/j.cca.2018.06.048. [DOI] [PubMed] [Google Scholar]
- Wan ES, Qiu W, Baccarelli A, Carey VJ, Bacherman H, Rennard SI, et al. 2012. Cigarette smoking behaviors and time since quitting are associated with differential DNA methylation across the human genome. Hum Mol Genet 21(13):3073–3082, PMID: 22492999, 10.1093/hmg/dds135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- White AJ, D’Aloisio AA, Nichols HB, DeRoo LA, Sandler DP. 2017. Breast cancer and exposure to tobacco smoke during potential windows of susceptibility. Cancer Causes Control 28(7):667–675, PMID: 28523418, 10.1007/s10552-017-0903-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wiklund P, Karhunen V, Richmond RC, Parmar P, Rodriguez A, De Silva M, et al. 2019. DNA methylation links prenatal smoking exposure to later life health outcomes in offspring. Clin Epigenet 11(1):97, PMID: 31262328, 10.1186/s13148-019-0683-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Williams SR, Aldred MA, Der Kaloustian VM, Halal F, Gowans G, McLeod DR, et al. 2010. Haploinsufficiency of HDAC4 causes brachydactyly mental retardation syndrome, with brachydactyly type E, developmental delays, and behavioral problems. Am J Hum Genet 87(2):219–228, PMID: 20691407, 10.1016/j.ajhg.2010.07.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xie C-M, Sun Y. 2019. The MTORC1-mediated autophagy is regulated by the FBXW7-SHOC2-RPTOR axis. Autophagy 15(8):1470–1472, PMID: 31010381, 10.1080/15548627.2019.1609864. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xinh PT, Tri NK, Nagao H, Nakazato H, Taketazu F, Fujisawa S, et al. 2003. Breakpoints at 1p36.3 in three MDS/AML(M4) patients with t(1;3)(p36;q21) occur in the first intron and in the 5′ region of MEL1. Genes Chromosomes Cancer 36(3):313–316, PMID: 12557231, 10.1002/gcc.10176. [DOI] [PubMed] [Google Scholar]
- Xu X, Balsiger R, Tyrrell J, Boyaka PN, Tarran R, Cormet-Boyaka E. 2015. Cigarette smoke exposure reveals a novel role for the MEK/ERK1/2 MAPK pathway in regulation of CFTR. Biochim Biophys Acta 1850(6):1224–1232, PMID: 25697727, 10.1016/j.bbagen.2015.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yamagata M, Weiner JA, Sanes JR. 2002. Sidekicks: synaptic adhesion molecules that promote lamina-specific connectivity in the retina. Cell 110(5):649–660, PMID: 12230981, 10.1016/S0092-8674(02)00910-8. [DOI] [PubMed] [Google Scholar]
- Yang AW, Sachs AJ, Nystuen AM. 2015. Deletion of Inpp5a causes ataxia and cerebellar degeneration in mice. Neurogenetics 16(4):277–285, PMID: 26051944, 10.1007/s10048-015-0450-4. [DOI] [PubMed] [Google Scholar]
- Yang N, Tian J, Wang X, Mei S, Zou D, Peng X, et al. 2020. A functional variant in TNXB promoter associates with the risk of esophageal squamous-cell carcinoma. Mol Carcinog 59(4):439–446, PMID: 32056283, 10.1002/mc.23166. [DOI] [PubMed] [Google Scholar]
- Zaghlool SB, Al-Shafai M, Al Muftah WA, Kumar P, Falchi M, Suhre K. 2015. Association of DNA methylation with age, gender, and smoking in an Arab population. Clin Epigenetics 7(1):6, PMID: 25663950, 10.1186/s13148-014-0040-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zeilinger S, Kühnel B, Klopp N, Baurecht H, Kleinschmidt A, Gieger C, et al. 2013. Tobacco smoking leads to extensive genome-wide changes in DNA methylation. PLoS One 8(5):e63812, PMID: 23691101, 10.1371/journal.pone.0063812. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang Y, Florath I, Saum K-U, Brenner H. 2016. Self-reported smoking, serum cotinine, and blood DNA methylation. Environ Res 146:395–403, PMID: 26826776, 10.1016/j.envres.2016.01.026. [DOI] [PubMed] [Google Scholar]
- Zhang B, Hong X, Ji H, Tang W-Y, Kimmel M, Ji Y, et al. 2018. Maternal smoking during pregnancy and cord blood DNA methylation: new insight on sex differences and effect modification by maternal folate levels. Epigenetics 13(5):505–518, PMID: 29945474, 10.1080/15592294.2018.1475978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhu X, Li J, Deng S, Yu K, Liu X, Deng Q, et al. 2016. Genome-wide analysis of DNA methylation and cigarette smoking in a Chinese population. Environ Health Perspect 124(7):966–973, PMID: 26756918, 10.1289/ehp.1509834. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ziller MJ, Gu H, Müller F, Donaghey J, Tsai LT-Y, Kohlbacher O, et al. 2013. Charting a dynamic DNA methylation landscape of the human genome. Nature 500(7463):477–481, PMID: 23925113, 10.1038/nature12433. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.