Skip to main content
Epigenetics Insights logoLink to Epigenetics Insights
. 2021 Mar 26;14:25168657211006159. doi: 10.1177/25168657211006159

Differential DNA Methylation by Hispanic Ethnicity Among Firefighters in the United States

Jaclyn M Goodrich 1,, Melissa A Furlong 2, Alberto J Caban-Martinez 3, Alesia M Jung 4, Ken Batai 5, Timothy Jenkins 6, Shawn Beitel 2, Sally Littau 2, John Gulotta 7, Darin Wallentine 7, Jeff Hughes 8, Charles Popp 9, Miriam M Calkins 10, Jefferey L Burgess 2
PMCID: PMC8756104  PMID: 35036834

Abstract

Firefighters are exposed to a variety of environmental hazards and are at increased risk for multiple cancers. There is evidence that risks differ by ethnicity, yet the biological or environmental differences underlying these differences are not known. DNA methylation is one type of epigenetic regulation that is altered in cancers. In this pilot study, we profiled DNA methylation with the Infinium MethylationEPIC in blood leukocytes from 31 Hispanic white and 163 non-Hispanic white firefighters. We compared DNA methylation (1) at 12 xenobiotic metabolizing genes and (2) at all loci on the array (>740 000), adjusting for confounders. Five of the xenobiotic metabolizing genes were differentially methylated at a raw P-value <.05 when comparing the 2 ethnic groups, yet were not statistically significant at a 5% false discovery rate (q-value <.05). In the epigenome-wide analysis, 76 loci exhibited DNA methylation differences at q < .05. Among these, 3 CpG sites in the promoter region of the biotransformation gene SULT1C2 had lower methylation in Hispanic compared to non-Hispanic firefighters. Other differentially methylated loci included genes that have been implicated in carcinogenesis in published studies (FOXK2, GYLTL1B, ZBTB16, ARHGEF10, and more). In this pilot study, we report differential DNA methylation between Hispanic and non-Hispanic firefighters in xenobiotic metabolism genes and other genes with functions related to cancer. Epigenetic susceptibility by ethnicity merits further study as this may alter risk for cancers linked to toxic exposures.

Keywords: Occupational health, epigenome-wide analysis study, xenobiotic metabolism, health disparities, occupational exposures

Introduction

Firefighters are a unique occupational group exposed to a variety of hazards including chemical exposures and physical and mental stressors. Exposure to combustion byproducts from fires is a major concern since a number are known or suspected human carcinogens.1-3 Firefighters have an increased overall risk of cancer incidence and mortality compared to the general population, as well as increased incidence and mortality for specific cancers, including those of the digestive tract, bladder, prostate, testicles, thyroid, and blood, among others.4-7

Ethnic and racial differences in firefighter cancer rates have been reported in the limited number of epidemiologic studies with a sufficient number of minority firefighters. In a cancer registry-based case control study in California, rates of 6 cancers (tongue, testicular, bladder, non-Hodgkin’s lymphoma, chronic leukocytic leukemia, and chronic myelogenous leukemia) were significantly elevated compared to the general population only in minority firefighters, most of whom were either Hispanic (62.2%) or black (27.7%). 7 In addition, non-minority and minority firefighters both had significantly elevated risks for 6 other cancers (melanoma, prostate, kidney, brain, multiple myeloma, and overall leukemia). In US civilian populations, adults of Hispanic ethnicity have been found to have a lower incidence of lung cancer, but an increase in cervical, gall bladder, liver, and gastric cancer8,9 compared to non-Hispanic whites.

The reasons for the observed cancer disparities among firefighters by race and ethnicity is not known. Potential explanations include but are not limited to differential occupational exposures, such as assignment to busier stations or differential environmental exposures (eg, from place of residence or diet). Of interest to this study, epigenetic or genetic differences between groups may also alter susceptibility to the impacts of exposures. Polycyclic aromatic hydrocarbons (PAHs) are a family of chemicals including known and suspected carcinogens which are produced during combustion. PAHs have a ubiquitous presence in the fire service, having been documented at the fireground during suppression, during overhaul activities, on firefighter personal protective equipment, and at fire stations.10-13 Additionally, biomonitoring studies have observed significant PAH exposures across various firefighters and fire service personnel after fire events, regardless of job assignment.2,14,15 Occupational exposure to mixtures of PAHs and other chemicals could contribute to the increased risk for lung, skin, and prostate cancers,16-18 which are also increased in firefighters compared to the general population.4,5,7,19

Epigenetic and genetic differences that alter the body’s ability to detoxify carcinogenic exposures, such as but not limited to PAHs, may modify susceptibility to cancers among firefighters with similar exposures. The epigenome consists of modifications to DNA and chromatin that do not alter the underlying DNA sequence yet are heritable, at least across cell divisions. DNA methylation is one major type of epigenetic regulation that is fairly stable across time and is typically associated with repression of gene expression. 20 It has been shown in adults to be responsive to a multitude of environmental factors including PAHs, other toxicant exposures,21-24 and stress. 25 We have previously shown differential blood DNA methylation in incumbent compared to new recruit firefighters, adjusted for age, and other covariates, as well as differential whole blood microRNA expression.26,27 DNA methylation has been shown to vary based on sex,28,29 race, 30 and ethnicity 31 at many genes. This may be due to differences in the underlying genetic sequence32,33 and/or disproportionate exposures to toxicants or psychosocial stressors. In particular, one cross-sectional study showed that over 70% of the variance in DNA methylation between ethnic groups was explained by shared genomic ancestry. 34 Interestingly, they identified that CpG sites that were previously reported to be responsive to exogenous exposures were enriched among the loci associated with ethnicity. 34 Additionally, although exposures to smoking and other combustion byproducts, 35 race/ethnicity, 8 and the epigenome36-38 have all been associated with cancer, whether and how the intersection of the 3 relates to cancer risk is unclear.

In this pilot study, we profiled and compared DNA methylation in white firefighters of Hispanic (n = 31) and non-Hispanic (n = 163) ethnicity. We hypothesized that genes differentially methylated between the ethnic groups would have functions relevant to carcinogenesis. We quantified DNA methylation at >850 000 cytosine-guanine dinucleotides (CpG sites) via the Infinium MethylationEPIC 39 in blood leukocyte DNA. We utilized a two-tiered analysis approach. We first compared DNA methylation levels in 12 genes relevant to metabolism and detoxification of PAHs—exposures among firefighters that are known to be carcinogenic. Second, we compared DNA methylation at all CpG sites included on the EPIC array to discover genes in other pathways. We then report on genes that are differentially methylated according to ethnicity.

Methods

Cohort recruitment and study population

This study included participants from 2 larger cancer prevention studies: the first a 3-year research project working in partnership with the Tucson Fire Department (TFD) and the second the prospective multicenter Fire Fighter Cancer Cohort Study (FFCCS) involving multiple fire departments and universities as well as the National Institute for Occupational Safety and Health (NIOSH). Firefighters were recruited for this analysis from 5 fire departments in Arizona (from 2016 to 2018), California (2019), and Massachusetts (2018), 2 of which were volunteer fire departments. Inclusion criteria for enrollment in the current study included being an active duty firefighter (including emergency medical responder) responding to fires as part of normal duties. All study procedures were approved by the institutional review boards (IRB) of the University of Arizona (IRB approval No. 1509137073) and the University of Miami (IRB approval No. 20170997).

The research team delivered an in-depth explanation of the study design, including potential risks and responsibilities, before all subjects provided informed consent. The survey questions involved standard demographic information plus general information such as previous cancer diagnosis, body weight, height, working duration as firefighters (including at current and previous departments), and tobacco use. Blood samples for DNA methylation measurements were collected during the day by qualified phlebotomists in one 6.0 ml dipotassium ethylene diaminetetraacetic acid (K2EDTA) tube (Beckton, Dickinson and Company, Franklin Lakes, NJ). Blood was stored frozen (temporarily at −20°C followed by long-term storage at −80°C) until use. 27

DNA methylation analysis

DNA was isolated from blood leukocytes. Concentration of double stranded DNA was measured via a QuantiFluor dsDNA System (Promega) or a Qubit Fluorometer (Thermo Scientific). DNA was bisulfite converted using Zymo kits. DNA methylation was quantified at >850 000 CpG sites throughout the genome using the Infinium MethylationEPIC array. 39 Samples were randomized and hybridized to chips during each batch, and personnel running the analysis were blinded to phenotype information about the samples (eg, ethnic group). Samples were run in 3 batches (one at the University of Utah DNA Sequencing and Genomics Core Facility, and 2 batches at the University of Michigan Advanced Genomics Core) and scanned by experienced personnel. Both Core facilities follow the same recommended protocol for the MethylationEPIC kit and scan the arrays using iScan instruments (Illumina, Inc). All batches included samples from both Hispanic and non-Hispanic firefighters. Raw image files were read with the R package minfi, 40 and quality control and normalization occurred using the package ENmix. 41 Quality control included comparing recorded sex to estimated sex based on signals on X and Y chromosomes, examining median intensities, bisulfite conversion efficiency, and more. Probes were removed if at least 5% of samples were not detected (P-value >1e−16 compared to background), and samples with >5% of probes not detected were excluded. Background correction was performed with noob and dye bias correction with Regression on Logarithm of Internal Control probes (RELIC). Quantile normalization was used to normalize intensities separately for methylated and unmethylated for type I and type II probes. 42 Probes that are known to be cross-reactive, have SNPs in the CpG or single-base extension site, or are on X and Y chromosomes were excluded. The final probe number used in downstream analyses covered 740 842 CpG sites; probes were annotated using the ilm10b4.h19 annotation R package. Analysis included 194 samples from white Hispanic or white non-Hispanic active firefighters with data passing all quality control.

The proportion of 6 cell types were estimated using reference data from sorted blood cells according to the algorithm established by Houseman et al.43,44 Surrogate variable analysis was performed using the intensity values from the non-negative control probes to create variables representing technical variation influencing the DNA methylation data. 45 Three principal components (PCs) from this analysis explained 92% of the technical variance in the data and were used as covariates in downstream models.

Statistical analysis

All data pre-processing and statistical analyses were conducted in the R Project for Statistical Computing (version ⩾3.6.3). Descriptive statistics were first calculated for all demographic variables and compared between Hispanic and non-Hispanic firefighters using t-tests or chi-square tests as appropriate. In all described analyses, data were only included from self-identified white Hispanic and white non-Hispanic individuals. Singular Value Decomposition (SVD) analysis was performed with the ChAMP package in R to identify technical and biological covariates that correlate with variation in the DNA methylation data. Briefly, the correlation between PCs of the methylation data with covariates was determined using linear regression for continuous variables or Kruskal-Wallis for categorical variables. 38 Covariates considered included age, gender, ethnicity, BMI, estimated cell type proportions, smoking history (categorized as never smoker, former smoking, and not reported), and PCs explaining technical variation from the SVD. Based on this analysis, gender, age, cell types (represented by proportion of neutrophils), and 3 PCs for technical variation were selected for statistical models along with the independent variable of interest (ethnicity). BMI was not associated with DNA methylation, but was slightly higher in the Hispanic group (Table 1) and is considered in an additional model. We also ran models including adjustment for smoking history or total years working as a firefighter.

Table 1.

Study population characteristics by ethnicity.

Hispanic Non-hispanic
n (%) Mean (SD) n (%) Mean (SD) P-value c
Males 27 (87.1) 145 (89.0) 1
Females 4 (12.9) 18 (11.0)
BMI (kg m−2) a 27.9 (3.6) 26.8 (3.5) .16
Age (y) 38.4 (9.6) 38.6 (9.8) .92
Years firefighting 13.6 (9.1) 15.2 (9.2) .37
Smoking history
 Never 23 (74.2) 122 (74.8) .99
 Past 5 (16.1) 25 (15.3)
 Not reported 3 (9.7) 16 (9.8)
Estimated cell type proportions (%)
 Neutrophils 58.9 (8.3) 57.2 (9.0) .32
 CD4+ T cells 12.7 (3.2) 13.8 (4.9) .11 b
 CD8+ T cells 10.0 (4.0) 10.6 (4.4) .51
 B cells 5.7 (2.4) 5.2 (2.4) .32
 Monocytes 7.5 (2.6) 8.0 (2.6) .34
 NK cells 6.3 (3.4) 6.1 (2.7) .83
a

Some subjects were missing BMI. Thirty Hispanic and 158 non-Hispanic had BMI. For all other variables there are 31 Hispanic and 163 non-Hispanic participants. All participants are white.

b

Welch’s t-test used due to unequal variances.

c

t-Test for continuous variables; chi square test for categorical variables.

We used a 2-tiered approach: (1) a hypothesis-driven investigation of genes known to influence response to environmental exposures with links to cancer risk; and (2) an exploratory approach investigating all loci available from the EPIC data. In the hypothesis driven approach, we included 187 CpG sites that annotated to 12 genes according to UCSC “refGene”: CYP1A1, CYP1B1, EPHX1, GSTA1, GSTM1, GSTP1, GSTT1, PTGS1, PTGS2, UGT1A1, UGT1A6, and UGT1A9. These genes were selected because they are involved in the metabolism and detoxification of PAHs, which are one set of exposures firefighters face in the line of duty. In the exploratory epigenome-wide approach, we included 740 842 CpG sites that passed quality control.

For the hypothesis-driven and exploratory approaches, linear regression was used to identify CpG sites differentially methylated between the ethnic groups. Models were fit to beta values (which represent the proportion methylated) for each CpG site separately. An empirical Bayes method in the limma R package46,47 was then used to shrink probe-wise variances towards a pooled estimate and calculate a moderated t-statistic prior to significance calling. For both approaches, CpG sites associated with ethnicity (Hispanic vs non-Hispanic) at a false discovery rate (FDR) adjusted P-value <.05 (referred to as q-value) are considered statistically significant. 48 Model 3 is considered the main model with all necessary potential confounders (eg, covariates associated with both DNA methylation and ethnicity). However, we also compared results with four other models (unadjusted, cell-free model, and models including additional adjustment for BMI, smoking history, or years firefighting):

  • Model 1 = ethnicity only (unadjusted model)

  • Model 2 = ethnicity + gender + age + PC1 + PC2 + PC3 (no-cell model)

  • Model 3 = Model 2 + Neutrophils (fully adjusted model)

  • Model 4 = Model 3 + BMI (n = 188 since some are missing BMI)

  • Model 5 = Model 3 + Smoking history

  • Model 6 = Model 3 + Years working as a firefighter

We performed a pathway analysis with the results from Model 3 to determine whether the top 10 000 CpG sites by raw P-value were enriched in certain biological or functional pathways. We used the gometh function in the missMethyl package, and ran pathway analysis separately for Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) terms. 49

Results

We profiled DNA methylation in blood leukocytes from 194 active US firefighters with an average of 15.0 (SD ± 9.2) total years of employment in the fire service. The firefighters were recruited from Massachusetts (n = 7), Southern California (n = 46), and Southern Arizona (n = 141). Of the 194, 188 were career firefighters and 6 (all from Arizona) were volunteer firefighters. Thirty-one of the firefighters included in this analysis identified as white Hispanic (16%) and the rest were white non-Hispanic. The average age was 38 years for both Hispanic and non-Hispanic firefighters. Smoking history, BMI, % female, years in the service, and estimated cell type proportions from the blood samples were similar among firefighters of both ethnicities (Table 1).

Hypothesis-driven approach

The EPIC array contained 187 CpG sites that annotated to 12 genes involved in PAH metabolism and detoxification. None of the CpG sites were associated with ethnicity using a 5% FDR significance cut-off (q < .05). Sixteen CpG sites in 5 of the genes were associated with ethnicity at an unadjusted P-value <.05 (Supplemental Table S1). These CpG sites, annotated to CYP1A1, CYP1B1, EPHX1, PTGS1, and UGT1A6 merit further exploration in larger multi-ethnic studies.

Epigenome-wide approach

In the main statistical model of all CpG sites, 54 had lower methylation in Hispanic firefighters compared to non-Hispanic firefighters and 22 had higher methylation (q < .05; Table 2). Effect sizes and significance values were similar in sensitivity analyses including in models without cell type adjustment and with adjustment for BMI, smoking history, or years working as a firefighter (Supplemental Table S2). Of the 76 significant loci, 21 were in intergenic regions and the remainder annotated to 46 genes. Effect estimates ranged from −15.2% lower in Hispanic firefighters (at chr1:210099448 in an intergenic region) to 19.1% higher (near the transcription start site of the CCDC66 gene; Table 2) compared with non-Hispanic participants. Two genes had 3 significant CpG sites each—FOXK2 and SULT1C2; the latter is a xenobiotic metabolizing enzyme (Figure 1). Five genes had 2 significant CpG sites each—ZBTB16, GYLTL1B, ARHGEF10, ZC2HC1A, and CRB2.

Table 2.

Differentially methylated CpG sites in Hispanic compared to non-Hispanic firefighters in the epigenome-wide analysis, fully adjusted model (q-value <.05).

ProbeID Position Gene a Effect estimate SE Average % methylation at CpG site P-value q-value
cg12545480 chr1: 210099448 NA −0.152 0.018 61.0 2.88E−15 2.13E−09
cg04800768 chr17: 80545544 FOXK2 −0.009 0.001 98.4 3.57E−12 1.32E−06
cg24582990 chr5: 154248355 CNOT8 −0.082 0.011 86.7 1.98E−11 4.88E−06
cg12289926 chr11: 68153810 LRP5 −0.050 0.008 83.3 2.77E−10 5.13E−05
cg26748898 chr17: 74912327 MGAT5B −0.079 0.013 70.8 2.90E−09 .000
cg04074945 chr11: 46071833 PHF21A 0.058 0.009 68.5 2.76E−09 .000
cg13227551 chr6: 144011222 PHACTR2 0.120 0.020 18.3 7.12E−09 .001
cg02796939 chr17: 80545125 FOXK2 −0.075 0.012 81.7 9.41E−09 .001
cg15386132 chr5: 133796338 NA 0.035 0.006 72.4 1.61E−08 .001
cg13968390 chr2: 108904812 SULT1C2 −0.138 0.024 77.2 1.90E−08 .001
cg12063639 chr9: 130184057 NA −0.135 0.024 91.4 4.80E−08 .003
cg04238311 chr11: 45945630 GYLTL1B −0.097 0.017 61.7 4.42E−08 .003
cg13962846 chr1: 7973298 NA −0.134 0.024 75.1 1.20E−07 .006
cg09513263 chr17: 70283945 NA −0.078 0.014 68.4 1.10E−07 .006
cg03142846 chr8: 1890946 ARHGEF10 −0.043 0.008 70.2 1.17E−07 .006
cg18912965 chr14: 91526996 RPS6KA5 −0.033 0.006 7.4 1.16E−07 .006
cg05194412 chr2: 137003533 NA −0.078 0.015 42.0 2.19E−07 .009
cg07846855 chr10: 77845945 C10orf11 −0.025 0.005 86.8 2.20E−07 .009
cg12648759 chr1: 161812851 ATF6 −0.034 0.006 82.7 2.57E−07 .010
cg05401945 chr3: 56590734 CCDC66 0.191 0.036 30.0 2.46E−07 .010
cg26905489 chr7: 83551804 NA −0.033 0.006 83.3 3.04E−07 .011
cg22139615 chr4: 141048272 MAML3 −0.065 0.012 84.1 3.33E−07 .011
cg23163573 chr2: 108905468 SULT1C2 −0.074 0.014 64.1 3.64E−07 .012
cg01297101 chr17: 37356023 RPL19 0.017 0.003 6.0 3.98E−07 .012
cg14720104 chr6: 118508739 SLC35F1 −0.064 0.012 36.6 4.40E−07 .013
cg12526471 chr11: 113929188 ZBTB16 −0.060 0.012 50.0 5.51E−07 .016
cg07737363 chr3: 53283930 TKT −0.036 0.007 17.2 6.53E−07 .018
cg01770799 chr16: 85951663 IRF8 −0.039 0.008 81.6 7.20E−07 .019
cg05468483 chr2: 70563880 NA −0.011 0.002 92.5 7.95E−07 .020
cg11031278 chr11: 113929114 ZBTB16 −0.061 0.012 58.0 1.08E−06 .025
cg03016722 chr9: 92517538 NA −0.053 0.011 66.4 1.10E−06 .025
cg24846680 chr1: 228362309 C1orf69 −0.020 0.004 96.4 1.07E−06 .025
cg14076011 chr20: 23312790 NA −0.069 0.014 82.2 1.16E−06 .026
cg02314394 chr5: 646139 CEP72 −0.066 0.013 92.6 1.23E−06 .026
cg24085707 chr17: 79615652 TSPAN10 −0.047 0.009 41.0 1.27E−06 .026
cg03582582 chr7: 139444053 HIPK2 0.038 0.008 67.5 1.25E−06 .026
cg21028142 chr17: 79581711 NPLOC4 0.098 0.020 66.7 1.29E−06 .026
cg04287289 chr16: 89883240 FANCA 0.021 0.004 1.3 1.33E−06 .026
cg20728173 chr3: 108791035 MORC1 0.055 0.011 49.3 1.36E−06 .026
cg08574105 chr9: 126137259 CRB2 −0.028 0.006 61.2 1.47E−06 .027
cg05945266 chr17: 80545020 FOXK2 −0.023 0.005 92.7 1.49E−06 .027
cg19662109 chr21: 45866122 NA −0.025 0.005 75.4 1.66E−06 .029
cg08544606 chr8: 1891006 ARHGEF10 −0.027 0.005 91.1 2.02E−06 .035
cg14487892 chr2: 64558493 NA −0.052 0.011 68.4 2.34E−06 .036
cg20674635 chr20: 44640803 MMP9 −0.036 0.007 16.3 2.40E−06 .036
cg16214826 chr2: 1065770 SNTG2 −0.025 0.005 92.7 2.25E−06 .036
cg24132527 chr5: 140019269 TMCO6 0.007 0.001 1.1 2.36E−06 .036
cg09353378 chr17: 34274396 NA 0.008 0.002 2.5 2.30E−06 .036
cg26245086 chr4: 87860957 AFF1 0.068 0.014 7.5 2.34E−06 .036
cg02786370 chr4: 2747928 TNIP2 −0.040 0.008 34.5 2.69E−06 .038
cg07162250 chr1: 7488166 CAMTA1 −0.040 0.008 54.9 2.58E−06 .038
cg02555772 chr16: 1079316 NA −0.031 0.006 93.6 2.68E−06 .038
cg03364549 chr20: 44622696 NA −0.030 0.006 91.8 2.72E−06 .038
cg16731079 chr21: 41758562 DSCAM 0.077 0.016 26.9 2.81E−06 .039
cg25838818 chr2: 108905173 SULT1C2 −0.103 0.021 39.6 2.87E−06 .039
cg14843888 chr3: 53530247 CACNA1D −0.033 0.007 69.8 3.13E−06 .041
cg21140145 chr8: 79577641 ZC2HC1A −0.098 0.021 32.6 3.28E−06 .042
cg00348244 chr17: 39222923 KRTAP2-4 0.010 0.002 93.6 3.37E−06 .042
cg13518537 chr8: 28618150 NA 0.076 0.016 39.0 3.33E−06 .042
cg07181209 chr5: 153585979 GALNT10 −0.083 0.017 60.6 3.44E−06 .042
cg02625222 chr9: 126135169 CRB2 −0.114 0.024 57.1 3.67E−06 .045
cg08655071 chr1: 209928895 TRAF3IP3 0.029 0.006 45.6 3.74E−06 .045
cg03983883 chr8: 79577618 ZC2HC1A −0.133 0.028 39.7 3.97E−06 .046
cg02524205 chr6: 167559851 NA 0.136 0.029 54.3 3.96E−06 .046
cg07370361 chr6: 36395612 PXT1 −0.065 0.014 66.6 4.18E−06 .047
cg24021129 chr5: 114408148 NA −0.014 0.003 88.0 4.12E−06 .047
cg20036516 chr17: 18006550 DRG2 0.017 0.004 86.4 4.22E−06 .047
cg23631229 chr1: 7962850 NA 0.004 0.001 1.2 4.35E−06 .047
cg23320965 chr2: 202850601 NA −0.095 0.020 69.7 4.68E−06 .048
cg27274564 chr2: 55534947 CCDC88A −0.054 0.011 92.0 4.66E−06 .048
cg24204282 chr11: 45944920 GYLTL1B −0.039 0.008 9.5 4.69E−06 .048
cg14023573 chr8: 140971379 TRAPPC9 0.017 0.004 88.0 4.63E−06 .048
cg17079172 chr9: 130265152 LRSAM1 0.023 0.005 79.2 4.50E−06 .048
cg09373597 chr3: 138667775 C3orf72 −0.041 0.009 41.7 4.97E−06 .049
cg15551881 chr9: 123688715 TRAF1 0.053 0.011 19.8 4.97E−06 .049
cg21803548 chr3: 144483077 NA −0.034 0.007 81.6 5.09E−06 .050

Effect estimates represent the change in proportion methylated in Hispanics (n = 31; compared to non-Hispanics, n = 163). Models adjusted for technical variation, age, gender, and estimated neutrophils.

a

NA means the CpG site is not within a gene or within a known feature (eg, promoter) of a specific gene.

Figure 1.

Figure 1.

DNA methylation at SULT1C2. Three CpG sites near the transcription start site (TSS) of SULT1C2 (probe IDs cg13968390, cg25838818, and cg23163573) had lower blood leukocyte DNA methylation in Hispanic compared with non-Hispanic firefighters. They were statistically significant after adjustment for gender, age, technical variation, and proportion of neutrophils (q-values = .001, .04, and .01). Average DNA methylation for Hispanic (green triangles) and non-Hispanic (blue squares) firefighters at the significant loci (labeled with *) and neighboring CpG sites covered on the EPIC array in SULT1C2 are shown.

In the pathway analysis, the top 25 pathways (P < .005) by raw P-value for GO and also 5 KEGG pathways with raw P-value <.05 are shown in Supplemental Table S3. Processes and biological functions represented in these pathways included cytoskeleton organization and actin binding, sensory perception, ossification, alanine transport, other metabolic-related pathways, and circadian rhythms. However, no concepts were statistically significant using a 5% FDR.

Discussion

In this pilot study of firefighters recruited from 3 US states, we observed differences in DNA methylation comparing Hispanic to non-Hispanic participants at 76 loci, including in a gene involved in xenobiotic metabolism and other genes that may be related to risk for carcinogenesis. These results provide preliminary evidence for an epigenetic mechanism underlying differential cancer risks seen in minority firefighters. In this study, no CpG sites from among 12 xenobiotic metabolizing genes were significantly different at a FDR of 5% in the hypothesis-driven approach. In the exploratory approach, 76 differentially methylated CpG sites by ethnicity were identified (q < .05). These sites were in genes that can activate chemical exposures (SULT1C2) 50 and genes that have been linked to cancer or cancer-related processes in other studies (FOXK2, GYLTL1B, ZBTB16, MAML3, RPS6KA5, ATF6, ZC2HC1A, and ARHGEF10).51-58

In the hypothesis-driven approach we focused on genes encoding biotransformation enzymes that are involved in handling chemical exposures to which firefighters are exposed. Cytochrome P450, epoxide hydrolase 1, glutathione-S-transferase, and prostaglandin-endoperoxide synthase are enzymes involved in PAH metabolism and detoxification, and variants and expression of the genes encoding these enzymes are associated with various types of cancer.18,59,60 In our study, there was no strong evidence for DNA methylation differences at these genes by ethnicity. However, CpG sites within CYP1A1, CYP1B1, EPHX1, PTGS1, and UGT1A6 demonstrated differential methylation at a raw P-value <.05, and should be explored further in larger studies. After activation by cytochrome P450 enzymes, PAH metabolites can bind to DNA and form DNA adducts; this damage can have a carcinogenic effect. 61 These genes may be dysregulated in minority firefighters more often compared to non-minority firefighters, and DNA adduct levels may increase risk for developing cancer at different rates between racial and ethnic groups. For example, although there was no difference in PAH-DNA adduct levels between African American and European American prostate cancer patients, 62 one study showed elevated DNA adduct levels were significantly associated with prostate cancer only in African Americans. 63

In the epigenome-wide analysis, most statistically significant CpG sites had decreased methylation in Hispanic compared to non-Hispanic firefighters. Three CpG sites with decreased methylation were in a CpG island within the gene body of FOXK2, a transcriptional regulator involved in glucose metabolism and autophagy. FOXK2 also has an emerging role in cancer. 51 DNA methylation at 3 CpG sites near the transcription start site of SULT1C2 was 7% to 14% lower in Hispanic compared to non-Hispanic firefighters (probe IDs cg13968390, cg25838818, and cg23163573; Figure 1). SULT1C2 was the only xenobiotic metabolizing gene to be identified in the epigenome-wide approach. This gene encodes an enzyme that activates carcinogenic hydroxylamines (such as N-hydroxy-2-acetylamino-fluorene). In an in vitro study, DNA methylation at one of the same CpG sites (cg13968390) was shown to inversely correlate with gene expression in a lung cell line. 50 Thus, reduced DNA methylation at this locus may increase SULT1C2 expression in Hispanic firefighters, which would increase the ability of the encoded enzyme to activate its substrates, including carcinogenic chemicals. Importantly, one of the differentially methylated CpG sites is also within an aryl hydrocarbon receptor (AhR) binding region, and AhR activation by exposures is thought to be a mechanism that promotes maintenance of hypomethylation in this region. 50 Thus, all firefighters would be expected to have reduced methylation at this AhR-binding region of SULT1C2 and increased expression due to AhR activation by occupational exposures (eg, PAHs; Figure 2). If Hispanic firefighters have a lower baseline methylation at this site to begin with due to genetic differences32-34 or other environmental exposures, this may further increase their risk for SULT1C2-related pathogenesis. Genetic polymorphisms in several sulfotransferase family genes are associated with cancers for which firefighters have elevated rates compared to the general population. 64 For SULT1C2, a polymorphism in the gene is associated with response to therapy in prostate cancer patients. 65

Figure 2.

Figure 2.

DNA methylation, occupational exposures, and disease risk: SULT1C2 as an example. DNA methylation at 3 CpG sites near the transcription start site of SULT1C2 was lower in Hispanic compared to non-Hispanic firefighters (1). Whether this is due to differences in genetics or life course environmental exposures or (2) due to differences in occupational exposures is not known. One of the differentially methylated CpG sites in SULT1C2 is within an AhR binding region. Activation of the AhR by occupational exposures (eg, PAHs) can also reduce methylation further. In an in vitro study, DNA methylation at the same CpG site inversely correlated with gene expression. 50 (3) Reduced DNA methylation at this gene may increase expression in Hispanic firefighters, which would increase the ability of SULT1C2 to activate carcinogenic exposures (4). Overall, these differences may lead to carcinogenesis later in life (5). In the figure: DNAm = DNA methylation; AhR = aryl hydrocarbon receptor; solid lines represent relationships with evidence from this or other studies; dashed lines represent hypothesized relationships.

We previously found AhR activation in dermal wipes and urine collected from firefighters following post-fire activities. The urinary AhR activity was somewhat correlated with urinary PAH metabolites, but also seems to be related to other unmeasured exposures, including other dioxin-like compounds. 66 In addition to the observation that the AhR binding region may be relevant for SULT1C2 methylation in our study sample, a large body of evidence implicates AhR as an important regulator of other genes that we report in Supplemental Table S1, including CYP1A1 67 and CYP1B1,68,69 both dioxin-inducible genes that are responsible for detoxification of a wide range of xenobiotic compounds. Interactions between genetics, epigenetics, and environmental exposures at genes like these may play a role in cancer. For example, an in vitro study showed that prostate cancer is associated with differential methylation of CYP1A1, and that the chromatic structure of this gene influences how it responds to dioxin exposure. 70 In another example, there was an interaction between genetic ancestry and smoking on DNA methylation levels in lung sputum with implications for lung cancer.71,72

Among genes annotated to CpG sites that differed by ethnicity, several of the genes have been implicated in carcinogenesis. Two CpG sites in the promoter region of ZBTB16 had 6% lower methylation in Hispanic firefighters compared to non-Hispanic. ZBTB16 encodes the promyelocytic leukemia zinc finger protein (PLZF), a transcription factor that has been implicated in acute childhood leukemia. 73 Whether PLZF plays a role in chronic leukemia, including leukemias that minority firefighters are at a higher risk for, is currently unknown. Recent studies suggest that ZBTB16 expression also inhibits metastasis of breast and prostate cancers.55,58 Two CpG sites in the gene body of GYLTL1B had lower methylation (4% and 10%) in Hispanic firefighters. This gene, also known as LARGE2, is a glycosyltransferase that has been implicated in many cancers including chronic lymphocytic leukemia (which is increased in minority firefighters), 7 renal cell carcinoma, and prostate cancer.56,57,74,75

Among the differentially methylated genes, RPS6KA5 is a serine/threonine-protein kinase that is important for transcriptional regulation, including for proto-oncogenes c-fos/FOS and c-jun/JUN. 53 A CpG site near the transcription start site of RPS6KA5 has 3.3% lower methylation in Hispanic firefighters (q = .006). ATF6 is another transcription factor that may play a role in survival of quiescent proliferative squamous carcinoma cells, and a CpG site in the gene body of ATF6 has 3.4% lower methylation in Hispanic compared to non-Hispanic firefighters (q = .01). Two CpG sites in ARHGEF10 had lower methylation in Hispanic firefighters. ARHGEF10 functions as a tumor suppressor, 52 and the methylation status of this gene may be responsive to environmental exposures. 76 ZC2HC1A expression has been implicated in an in vitro study of cancer, 54 and 2 CpG sites in this gene had lower methylation among Hispanic firefighters.

This study had several strengths and limitations. To assess differences in DNA methylation by Hispanic compared to non-Hispanic ethnicity, we leveraged samples from a cohort of firefighters. The unique study sample, which consisted of healthy white firefighters, enabled us to reduce confounding by occupation, occupational exposures, age, race, health status, and socioeconomic status. Our two-tiered analysis plan included a hypothesis-driven approach and identification of potential new targets via an epigenome-wide approach. While the total sample size was 194, only 16% were Hispanic, and thus statistical power was limited to detect all true associations after correcting for multiple comparisons. We report associations with a q-value <.05, which is a commonly used method to account for multiple testing in epigenome-wide studies. The top 12 loci in the exploratory analysis also remain significant at a P-value <9.4 × 10−8, which is an alternative cut-off recommended for EPIC data that considers the number of total sites and their intra-individual correlation. 77 We acknowledge that “Hispanic” is a broad classification that encompasses many ethnic groups and varying genetic ancestry 34 ; results should be followed up in larger cohorts with more granular classification of ethnicity and race and genetic data to estimate ancestry. The majority of study participants were male, and sex-specific differences in DNA methylation by ethnicity could not be assessed due to the sample size. Since the study is cross-sectional, we are uncertain if the differences in DNA methylation by ethnicity predated the study participants’ firefighting exposures, and/or if their exposures contributed to the observed differences. We lacked a control group of non-firefighters to help address this uncertainty. However, adjusting for years firefighting as a crude proxy of exposure did not change the results. Since exposure assessment (eg, for PAHs, etc.) was not performed, we cannot exclude the possibility that Hispanic and non-Hispanic firefighters were unequally exposed to epigenetic-modifying toxicants or stressors in the workplace. Environmental conditions in their past 78 or present residential environments could also contribute to the differences seen here.

Conclusion

In summary, this pilot study provides preliminary evidence for differences in DNA methylation by ethnicity, including in genes relevant to carcinogenesis, among firefighters who are at increased risk for multiple cancers due to occupational exposures. In the epigenome-wide approach, we report differential methylation at 76 CpG sites by ethnicity (q < .05). Some of the differentially methylated loci are in genes that have functions related to carcinogenesis (eg, GYLTL1B, ZBTB16, ARHGEF10, and RPS6KA5) or xenobiotic metabolism (SULT1C2). Given the increased risk firefighters already have for multiple cancers due to hazardous occupational exposures, it is imperative that we understand additional risk factors, including epigenetic susceptibility, that may alter this long-term risk among all firefighters and also specifically among ethnic or racial groups. Ultimately, the incorporation of such information into environmental or occupational risk assessment should be used to protect the most susceptible individuals, even within worker groups.

Supplemental Material

sj-xlsx-1-gae-10.1177_25168657211006159 – Supplemental material for Differential DNA Methylation by Hispanic Ethnicity Among Firefighters in the United States

Supplemental material, sj-xlsx-1-gae-10.1177_25168657211006159 for Differential DNA Methylation by Hispanic Ethnicity Among Firefighters in the United States by Jaclyn M Goodrich, Melissa A Furlong, Alberto J Caban-Martinez, Alesia M Jung, Ken Batai, Timothy Jenkins, Shawn Beitel, Sally Littau, John Gulotta, Darin Wallentine, Jeff Hughes, Charles Popp, Miriam M Calkins and Jefferey L Burgess in Epigenetics Insights

Acknowledgments

We first and foremost acknowledge the firefighter participants for graciously volunteering their time and samples to be in the study. We also acknowledge the partnering fire departments and their locals for their contribution to this research. We acknowledge Alisa Dewald at the University of Michigan for sample preparation. The University of Michigan Advanced Genomics Core and the University of Utah DNA Sequencing and Genomics Core Facility completed the EPIC analyses.

Footnotes

Funding: The author(s) disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This research was supported by the National Institute of Environmental Health Sciences (NIEHS), specifically by supplements to the P30 Centers at the University of Arizona (Grant No. P30 ES006694) and the University of Michigan (Grant No. P30 ES017885). MF was supported by NIEHS Grant No. K99ES028743. The US Federal Emergency Management Agency (FEMA) also supported this work (Grant Nos. EMW-2014-FP-00200, EMW-2015-FP-00213, and EMW-2018-FP-00086). The findings and conclusions in this paper are those of the authors and do not necessarily represent the official position of the National Institute for Occupational Safety and Health (NIOSH), Centers for Disease Control and Prevention. Mention of trade names and commercial products does not constitute endorsement or recommendation for use by NIOSH.

Declaration of conflicting interests: The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Authors Contribution: JMG, JLB, and AJCM designed the research study; JLB, AJCM, and MMC obtained funding and designed protocols to develop the cohorts; JG, DW, JH, and CP recruited subjects and collected data; AMJ, SB, SL, MMC, and JLB managed data and samples; JMG and TJ conducted the DNA methylation analysis; JMG conducted statistical analysis and drafted the manuscript; and MAF, KB, TJ, MMC, and JLB provided interpretation of results. All authors contributed to the writing and/or editing of the manuscript.

ORCID iDs: Jaclyn M Goodrich Inline graphic https://orcid.org/0000-0002-8289-9253

Alberto J Caban-Martinez Inline graphic https://orcid.org/0000-0002-5960-1308

Supplemental material: Supplemental material for this article is available online.

References

  • 1. Bolstad-Johnson DM, Burgess JL, Crutchfield CD, Storment S, Gerkin R, Wilson JR. Characterization of firefighter exposures during fire overhaul. Aihaj. 2000;61:636-641. [DOI] [PubMed] [Google Scholar]
  • 2. Fent KW, Toennis C, Sammons D, et al. Firefighters’ absorption of PAHs and VOCs during controlled residential fires by job assignment and fire attack tactic. J Expo Sci Environ Epidemiol. 2020;30:338-349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. IARC Working Group on the Evaluation of Carcinogenic Risks to Humans. Painting, firefighting, and shiftwork. In IARC Monographs on the Evalulation of Carcinogenic Risks to Humans, no 98. Lyon (FR): International Agency for Research on Cancer; (IARC); 2010. [PMC free article] [PubMed] [Google Scholar]
  • 4. Daniels RD, Kubale TL, Yiin JH, et al. Mortality and cancer incidence in a pooled cohort of US firefighters from San Francisco, Chicago and Philadelphia (1950-2009). Occup Environ Med. 2014;71:388-397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Jalilian H, Ziaei M, Weiderpass E, Rueegg CS, Khosravi Y, Kjaerheim K. Cancer incidence and mortality among firefighters. Int J Cancer. 2019;145:2639-2646. [DOI] [PubMed] [Google Scholar]
  • 6. LeMasters GK, Genaidy AM, Succop P, et al. Cancer risk among firefighters: a review and meta-analysis of 32 studies. J Occup Environ Med. 2006;48:1189-1202. [DOI] [PubMed] [Google Scholar]
  • 7. Tsai RJ, Luckhaupt SE, Schumacher P, Cress RD, Deapen DM, Calvert GM. Risk of cancer among firefighters in California, 1988-2007. Am J Ind Med. 2015;58:715-729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Haile RW, John EM, Levine AJ, et al. A review of cancer in US Hispanic populations. Cancer Prev Res. 2012;5:150-163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Miller KD, Sauer AG, Ortiz AP, et al. Cancer statistics for hispanics/latinos, 2018. CA Cancer J Clin. 2018;68:425-445. [DOI] [PubMed] [Google Scholar]
  • 10. Fent KW, Alexander B, Roberts J, et al. Contamination of firefighter personal protective equipment and skin and the effectiveness of decontamination procedures. J Occup Environ Hyg. 2017;14:801-814. [DOI] [PubMed] [Google Scholar]
  • 11. Fent KW, Eisenberg J, Snawder J, et al. Systemic exposure to PAHs and benzene in firefighters suppressing controlled structure fires. Ann Occup Hyg. 2014;58:830-845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Baxter CS, Hoffman JD, Knipp MJ, Reponen T, Haynes EN, et al. Exposure of firefighters to particulates and polycyclic aromatic hydrocarbons. J Occup Environ Hyg. 2014;11:D85-D91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Stec AA, Dickens KE, Salden M, et al. Occupational exposure to polycyclic aromatic hydrocarbons and elevated cancer incidence in firefighters. Sci Rep. 2018;8:2476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Burgess JL, Hoppe-Jones C, Griffin SC, et al., Evaluation of interventions to reduce firefighter exposures. J Occup Environ Med. 2020;62:279-288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Laitinen J, Mäkelä M, Mikkola J, Huttu I. Fire fighting trainers’ exposure to carcinogenic agents in smoke diving simulators. Toxicol Lett. 2010;192:61-65. [DOI] [PubMed] [Google Scholar]
  • 16. Stenehjem JS, Robsahm TE, Bråtveit M, Samuelsen SO, Kirkeleit J, Grimsrud TK, et al. Aromatic hydrocarbons and risk of skin cancer by anatomical site in 25 000 male offshore petroleum workers. Am J Ind Med. 2017;60:679-688. [DOI] [PubMed] [Google Scholar]
  • 17. Armstrong B, Hutchinson E, Unwin J, Fletcher T. Lung cancer risk after exposure to polycyclic aromatic hydrocarbons: a review and meta-analysis. Environ Health Perspect. 2004;112:970-978. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Rybicki BA, Nock NL, Savera AT, Tang D, Rundle A. Polycyclic aromatic hydrocarbon-DNA adduct formation in prostate carcinogenesis. Cancer Lett. 2006;239:157-167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Lee DJ, Koru-Sengul T, Hernandez MN, et al. Cancer risk among career male and female Florida firefighters: evidence from the Florida Firefighter Cancer Registry (1981-2014). Am J Ind Med. 2020;63:285-299. [DOI] [PubMed] [Google Scholar]
  • 20. Jones PA. Functions of DNA methylation: islands, start sites, gene bodies and beyond. Nat Rev Genet. 2012;13:484-492. [DOI] [PubMed] [Google Scholar]
  • 21. Furlong MA, Paul KC, Yan Q, et al. An epigenome-wide association study of ambient pyrethroid pesticide exposures in California’s central valley. Int J Hyg Environ Health. 2020;229:113569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Li J, Zhu X, Yu K, et al. Exposure to polycyclic aromatic hydrocarbons and accelerated DNA methylation aging. Environ Health Perspect. 2018;126:067005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Kim YH, Lee YS, Lee DH, Kim DS, et al. Polycyclic aromatic hydrocarbons are associated with insulin receptor substrate 2 methylation in adipose tissues of Korean women. Environ Res. 2016;150:47-51. [DOI] [PubMed] [Google Scholar]
  • 24. Dai L, Mehta A, Mordukhovich I, et al., Differential DNA methylation and PM2.5 species in a 450K epigenome-wide association study. Epigenetics. 2017;12:139-148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Lam LL, Emberly E, Fraser HB, et al. Factors underlying variable DNA methylation in a human community cohort. Proc Natl Acad Sci USA. 2012;109:17253-17260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Jeong KS, Zhou J, Griffin SC, et al. MicroRNA changes in firefighters. J Occup Environ Med. 2018;60:469-474. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Zhou J, Jenkins TG, Jung AM, et al. DNA methylation among firefighters. PLoS One. 2019;14:e0214282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Zhang B, Hong X, Ji H, et al. Maternal smoking during pregnancy and cord blood DNA methylation: new insight on sex differences and effect modification by maternal folate levels. Epigenetics. 2018;13:505-518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. McCarthy MM, Auger AP, Bale TL, et al. The epigenetics of sex differences in the brain. J Neurosci. 2009;29:12815-12823. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Adkins RM, Krushkal J, Tylavsky FA, Thomas F. Racial differences in gene-specific DNA methylation levels are present at birth. Birth Defects Res A: Clin Mol Teratol. 2011;91:728-736. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Park SL, Patel YM, Loo LWM, et al. Association of internal smoking dose with blood DNA methylation in three racial/ethnic populations. Clin Epigenetics. 2018;10:110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. McKennan C, Naughton K, Stanhope C, et al. Longitudinal data reveal strong genetic and weak non-genetic components of ethnicity-dependent blood DNA methylation levels. Epigenetics. 2020. doi: 10.1080/15592294.2020.1817290 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Bell JT, Pai AA, Pickrell JK, et al. DNA methylation patterns associate with genetic and gene expression variation in HapMap cell lines. Genome Biol. 2011;12:R10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Galanter JM, Gignoux CR, Oh SS, et al. Differential methylation between ethnic sub-groups reflects the effect of genetic ancestry and environmental exposures. Elife. 2017;6:e20532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Sasco A, Secretan M, Straif K. Tobacco smoking and cancer: a brief review of recent epidemiological evidence. Lung Cancer. 2004;45:S3-S9. [DOI] [PubMed] [Google Scholar]
  • 36. Yi JM, Dhir M, Neste LV, et al. Genomic and epigenomic integration identifies a prognostic signature in colon cancer. Clin Cancer Res. 2011;17:1535-1545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Widschwendter M, Jones A, Evans I, et al. Epigenome-based cancer risk prediction: rationale, opportunities and challenges. Nat Rev Clin Oncol. 2018;15:292. [DOI] [PubMed] [Google Scholar]
  • 38. Teschendorff AE, Menon U, Gentry-Maharaj A, et al. An epigenetic signature in peripheral blood predicts active ovarian cancer. PLoS One. 2009;4:e8274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Moran S, Arribas C, Esteller M. Validation of a DNA methylation microarray for 850,000 CpG sites of the human genome enriched in enhancer sequences. Epigenomics. 2016;8:389-399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Fortin JP, Triche TJ, Jr, Hansen KD. Preprocessing, normalization and integration of the Illumina HumanMethylationEPIC array with minfi. Bioinformatics. 2016;33:558-560. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Xu Z, Langie SAS, De Boever P, Taylor JA, Niu L, et al. RELIC: a novel dye-bias correction method for Illumina Methylation BeadChip. BMC Genomics. 2017;18:4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Pidsley R, Zotenko E, Peters TJ, et al. Critical evaluation of the illumina methylationepic beadchip microarray for whole-genome DNA methylation profiling. Genome Biol. 2016;17:208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Houseman EA, Accomando WP, Koestler DC, et al. DNA methylation arrays as surrogate measures of cell mixture distribution. BMC Bioinformatics. 2012;13:86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Salas LA, Koestler DC, Butler RA, et al. An optimized library for reference-based deconvolution of whole-blood biospecimens assayed using the Illumina HumanMethylationEPIC BeadArray. Genome Biol. 2018;19:64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Teschendorff AE, Zhuang J, Widschwendter M. Independent surrogate variable analysis to deconvolve confounding factors in large-scale microarray profiling studies. Bioinformatics. 2011;27:1496-1505. [DOI] [PubMed] [Google Scholar]
  • 46. Ritchie ME, Phipson B, Wu D, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43:e47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Smyth GK. Limma: Linear models for microarray data. In: Gentleman R., et al., eds. Bioinformatics and Computational Biology Solutions using R and Bioconductor, R. New York; 2005:397-420. [Google Scholar]
  • 48. Hochberg Y, Benjamini Y. More powerful procedures for multiple significance testing. Stat Med. 1990;9:811-818. [DOI] [PubMed] [Google Scholar]
  • 49. Phipson B, Maksimovic J, Oshlack A. missMethyl: an R package for analyzing data from Illumina’s HumanMethylation450 platform. Bioinformatics. 2016;32:286-288. [DOI] [PubMed] [Google Scholar]
  • 50. Johnson CJS. CpG Poor Promoter SULT1C2 Regulated by DNA Methylation is Induced by Cigarette Smoke Condensate in Lung Cell Lines. University of Southern California: ProQuest Dissertation Publishing; 2015. [Google Scholar]
  • 51. Nestal de Moraes G, da Torre Carneiro L, Maia RC, Wing-Fai Lam E, Sharrocks AD. FOXK2 transcription factor and its emerging roles in cancer. Cancers. 2019;11:393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Joseph J, Radulovich N, Wang T, Raghavan V, Zhu C-Q, Tsao M-S. Rho guanine nucleotide exchange factor ARHGEF10 is a putative tumor suppressor in pancreatic ductal adenocarcinoma. Oncogene. 2020;39:308-321. [DOI] [PubMed] [Google Scholar]
  • 53. Soloaga A, Thomson S, Wiggin GR, et al. MSK2 and MSK1 mediate the mitogen- and stress-induced phosphorylation of histone H3 and HMG-14. EMBO J. 2003;22:2788-2797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Zhang J, Cong R, Zhang Q, Zeng T, Song R, Meng X. Integrative analysis of ceRNA network and DNA methylation associated with gene expression in malignant pheochromocytomas: a study based on The Cancer Genome Atlas. Transl Androl Urol. 2020;9:344-354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. He J, Wu M, Xiong L, et al. BTB/POZ zinc finger protein ZBTB16 inhibits breast cancer proliferation and metastasis through upregulating ZBTB28 and antagonizing BCL6/ZBTB27. Clin Epigenetics. 2020;12:82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Esser AK, Miller MR, Huang Q, et al. Loss of LARGE2 disrupts functional glycosylation of α-dystroglycan in prostate cancer. J Biol Chem. 2013;288:2132-2142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Huang Q, Miller MR, Schappet J, Henry MD. The glycosyltransferase LARGE2 is repressed by Snail and ZEB1 in prostate cancer. Cancer Biol Ther. 2015;16:125-136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Xiao GQ, Unger P, Yang Q, et al. Loss of PLZF expression in prostate cancer by immunohistochemistry correlates with tumor aggressiveness and metastasis. PLoS One. 2015;10:e0121318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Ewa B, Danuta M. Polycyclic aromatic hydrocarbons and PAH-related DNA adducts. J Appl Genet. 2017;58:321-330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Wiese FW, Thompson PA, Kadlubar FF. Carcinogen substrate specificity of human COX-1 and COX-2. Carcinogenesis. 2001;22:5-10. [DOI] [PubMed] [Google Scholar]
  • 61. Barnes JL, Zubair M, John K, Poirier MC, Martin FL. Carcinogens and DNA damage. Biochem Soc Trans. 2018;46:1213-1224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Rybicki BA, Neslund-Dudas C, Bock CH, et al. Polycyclic aromatic hydrocarbon–DNA adducts in prostate and biochemical recurrence after prostatectomy. Clin Cancer Res. 2008;14:750-757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Tang D, Kryvenko ON, Wang Y, et al. Elevated polycyclic aromatic hydrocarbon-DNA adducts in benign prostate and risk of prostate cancer in African Americans. Carcinogenesis. 2013;34:113-120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Daniels J, Kadlubar S. Sulfotransferase genetic variation: from cancer risk to treatment response. Drug Metab Rev. 2013;45:415-422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Deeken JF, Cormier T, Price DK, et al. A pharmacogenetic study of docetaxel and thalidomide in patients with castration-resistant prostate cancer using the DMET genotyping platform. Pharmacogenomics J. 2010;10:191-199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Beitel SC, Flahr LM, Hoppe-Jones C, et al. Assessment of the toxicity of firefighter exposures using the PAH CALUX bioassay. Environ Int. 2020;135:105207. [DOI] [PubMed] [Google Scholar]
  • 67. Nebert DW, Roe AL, Dieter MZ, Solis WA, Yang Y, Dalton TP. Role of the aromatic hydrocarbon receptor and [Ah] gene battery in the oxidative stress response, cell cycle control, and apoptosis. Biochem Pharmacol. 2000;59:65-85. [DOI] [PubMed] [Google Scholar]
  • 68. Bock KW, Köhle C. Ah receptor: dioxin-mediated toxic responses as hints to deregulated physiologic functions. Biochem Pharmacol. 2006;72:393-404. [DOI] [PubMed] [Google Scholar]
  • 69. Helmig S, Seelinger JU, Döhre J, Schneider J. RNA expressions of AHR, ARNT and CYP1B1 are influenced by AHR Arg554Lys polymorphism. Mol Genet Metab. 2011;104:180-184. [DOI] [PubMed] [Google Scholar]
  • 70. Okino ST, Pookot D, Li L-C, et al. Epigenetic inactivation of the dioxin-responsive cytochrome P4501A1 gene in human prostate cancer. Cancer Res. 2006;66:7420-7428. [DOI] [PubMed] [Google Scholar]
  • 71. Leng S, Liu Y, Thomas CL, et al. Native American ancestry affects the risk for gene methylation in the lungs of Hispanic smokers from New Mexico. Am J Respir Crit Care Med. 2013;188:1110-1116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Leng S, Wu G, Klinge DM, et al. Gene methylation biomarkers in sputum as a classifier for lung cancer risk. Oncotarget. 2017;8:63978-63985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Wasim M, Carlet M, Mansha M., et al. PLZF/ZBTB16, a glucocorticoid response gene in acute lymphoblastic leukemia, interferes with glucocorticoid-induced apoptosis. J Steroid Biochem Mol Biol. 2010;120:218-227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Miller MR, Ma D, Schappet J, et al. Downregulation of dystroglycan glycosyltransferases LARGE2 and ISPD associate with increased mortality in clear cell renal cell carcinoma. Mol Cancer. 2015;14:141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Mosquera Orgueira A, Rodríguez BA, Díaz Arias JÁ, González Pérez MS, Bello López JL. New recurrent structural aberrations in the genome of chronic lymphocytic leukemia based on exome-sequencing data. Front Genet. 2019;10:854. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Mohanty A.F., et al., Infant sex-specific placental cadmium and DNA methylation associations. Environ Res. 2015;138:74-81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Mansell G, Gorrie-Stone TJ, Bao Y, et al. Guidance for DNA methylation studies: statistical insights from the Illumina EPIC array. BMC Genomics. 2019;20:366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Reuben A, Sugden K, Arseneault L, et al. Association of neighborhood disadvantage in childhood with DNA methylation in young adulthood. JAMA Netw Open. 2020;3:e206095. [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.

Supplementary Materials

sj-xlsx-1-gae-10.1177_25168657211006159 – Supplemental material for Differential DNA Methylation by Hispanic Ethnicity Among Firefighters in the United States

Supplemental material, sj-xlsx-1-gae-10.1177_25168657211006159 for Differential DNA Methylation by Hispanic Ethnicity Among Firefighters in the United States by Jaclyn M Goodrich, Melissa A Furlong, Alberto J Caban-Martinez, Alesia M Jung, Ken Batai, Timothy Jenkins, Shawn Beitel, Sally Littau, John Gulotta, Darin Wallentine, Jeff Hughes, Charles Popp, Miriam M Calkins and Jefferey L Burgess in Epigenetics Insights


Articles from Epigenetics Insights are provided here courtesy of SAGE Publications

RESOURCES