Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Aug 1.
Published in final edited form as: Hum Genet. 2013 Apr 19;132(8):943–953. doi: 10.1007/s00439-013-1306-3

BET1L and TNRC6B associate with uterine fibroid risk among European Americans

Todd L Edwards 1,2,3,4,*, Kara A Michels 1,2,5,*, Katherine E Hartmann 1,2,5,3, Digna R Velez Edwards 1,2,3,5
PMCID: PMC3715562  NIHMSID: NIHMS470570  PMID: 23604678

Abstract

Uterine fibroid (UFs) affect 77% of women by menopause and account for $9.4 billion in healthcare costs each year. Although UFs are heritable, genetic risk is poorly understood. The first genome-wide association study (GWAS) of UFs was recently performed in a Japanese population, with reported genome-wide significance for single nucleotide polymorphisms (SNPs) across three chromosomal regions. We tested these SNPs for association with UFs in U.S. cohorts. Women were enrolled in the Right from the Start (RFTS) cohort and the BioVU DNA repository. UF status in both cohorts was determined by pelvic imaging. We tested 65 candidate and haplotype-tagging SNPs for association with UFs presence using logistic regression in RFTS and the top three GWAS associated SNPs in BioVU. We also combined association results from both cohorts using meta-analysis. 1,086 European American (EA) cases and 1,549 controls were examined. Two SNP associations replicated (blocked early in transport 1 homolog[BET1L] rs2280543, RFTS-BioVU meta-odds ratio[OR]=0.67 95% confidence interval[CI] 0.38 to 0.96, Q=0.70, I=0, p=6.9×10-3; trinucleotide repeat containing 6B[TNRC6B] rs12484776, RFTS-BioVU meta-OR=1.21, 95% CI 1.07 to 1.35, Q=0.24, I=28.37, p=8.7×10-3). Meta-analyses combining evidence from RFTS, BioVU, and prior GWAS showed little heterogeneity in effect sizes across studies, with meta-p-values between 7.45×10-8 to 3.89×10-9, which were stronger than prior GWAS and supported associations observed for all previously identified loci. These data suggest common variants increase risk for UF in both EA and Japanese populations. However, further research is needed to assess the role of these genes across other racial groups.

Keywords: Uterine leiomyoma, fibroids, genetic epidemiology, polymorphism, women's health

INTRODUCTION

Uterine leiomyomata, or fibroids (UFs), are the most common female pelvic tumor. Prevalence estimates range from 20% to 77%, increasing with age up to menopause.(Cramer and Patel, 1990;Marshall et al., 1997;Vollenhoven, 1998) Known risk factors for UFs include African American (AA) race,(Baird et al., 2003;Cramer and Patel, 1990;Faerstein et al., 2001;Marshall et al., 1997;Ojeda, 1979) early age-at-menarche, (Dragomir et al., 2010;Faerstein et al., 2001;Lumbiganon et al., 1996;Marshall et al., 1998;Samadi et al., 1996;Wise et al., 2004) high body mass index (BMI),(Moore et al., 2008;Takeda et al., 2008) and increased age.(Baird et al., 2003) In addition, a protective effect for UFs has also been observed with higher parity, likely due to pregnancy-related hormonal and physical changes including postpartum uterine involution.(Baird and Dunson, 2003;Laughlin et al., 2010a;Laughlin et al., 2011)

Multiple lines of evidence have shown that UFs are influenced by genetic risk factors. First, UFs are highly heritable with evidence from twin-pair and familial aggregation studies.(Luoto et al., 2000;Treloar et al., 1992) Heritability studies of UFs in several European populations have observed that between 26 and 69% of UF risk is due to genetic factors.(Kurbanova et al., 1989;Luoto et al., 2000;Snieder et al., 1998) Further supporting a genetic contribution to risk are the observed racial disparities in UF age of onset, number, size, and lifetime incidence by menopause.(Baird et al., 2003) Genetic epidemiology studies to date have been largely limited to small-scale or single marker studies of steroid hormones, particularly estrogen, as it is potentially the most critical regulator of fibroid growth.(Flake et al., 2003) Also other growth factors,(Sozen and Arici, 2002) reproductive factors,(Parazzini et al., 1996) dysregulation of microRNAs,(Marsh et al., 2008) shortening of telomeres,(Bonatz et al., 1998) excessive production of disorganized extracellular matrix,(Malik et al., 2010;Sozen and Arici, 2002) and acquired chromosomal aberrations have been noted in UF studies.(El-Gharib and Elsobky, 2010)

Recently a genome-wide association study (GWAS) by Cha and colleagues appeared in Nature Genetics that examined risk for UFs among a population of Japanese women.(Cha et al., 2011) Eleven single nucleotide polymorphisms (SNPs) in three chromosomal regions (10q24.33, 11p15.5, and 22q13.1) associated with increased risk of UFs. The SNPs identified in the GWAS mapped to or nearby the genes STE20-like kinase (SLK), oligonucleotide/oligosaccharide-binding fold containing 1 (OBFC1), trinucleotide repeat containing 6B (TNRC6B), outer dense fiber of sperm tails 3 (ODF3), blocked early in transport 1 homolog (BET1L), resistance to inhibitors of cholinesterase 8 homolog A (RIC8A), and sirtuin 3 (SIRT3). The degree to which these findings generalize across racial groups is unknown, as they have not been replicated in non-Asian populations. Based on findings from other complex diseases, the same genetic factors may not explain UF risk across racial groups. Furthermore, since this is a clinical cohort is may be that these SNPs are more associated with severe and/or symptomatic fibroids. To date there are no published GWAS of UFs in U.S. populations.

In efforts to examine the three chromosome regions more comprehensively for an association with UF risk, we examined haplotype-tagging and index SNPs in a European American (EA) U.S. population rather than limiting our selection to only the previously studied variants. To conduct this analysis we used two cohorts of women, all of whom had pelvic imaging performed to detect the presence of UFs. Imaging is critical, because many women with UFs are asymptomatic and without imaging, studies may misclassify as many as 51% of women.(Baird et al., 2003;Myers et al., 2012) The primary goal of this study was to determine if gene variants within the previously associated gene regions associate with UF risk in an independent U.S. population.

MATERIALS AND METHODS

Study Populations

Right from the Start (RFTS)

RFTS is a community-based pregnancy cohort that enrolled study participants between 2001 and 2012. RFTS enrolled participants from Galveston, Texas; Memphis, Nashville, Knoxville, and Chattanooga, Tennessee; and the Research Triangle region (Raleigh, Durham, and Chapel Hill) in North Carolina. These analyses included RFTS participants who were 18 years or older and non-Hispanic EAs. As a part of participation, consent was obtained to review study participant medical records. Direct marketing and recruitment strategies have been previously described.(Promislow et al., 2004) The institutional review board (IRB) of Vanderbilt University, Nashville, Tennessee approved this study.

At enrollment, a research transvaginal ultrasound was conducted to assess embryonic development for the study pregnancy and to systematically examine the uterus for presence of UFs. The fibroid measurement protocol required three separate sets of measurements for each UF, with assessment of three perpendicular diameters: length, width, and depth. RFTS includes fibroids as small as 0.5 centimeters (cm) in maximum diameter.(Laughlin et al., 2009) Multiple still images of each UF with caliper markings of each diameter were recorded and a UF map was completed indicating the location and type of all UF(s).

Participants completed an intake interview at enrollment and a computer assisted telephone interview at the end of the first trimester. The intake and first trimester interviews provided information on reproductive history and candidate confounders. DNA samples were obtained either in person or by mail during follow-up using Oragene saliva DNA kits (DNA Genotek Inc., Ontario, Canada).

The BioVU DNA Repository

The BioVU Repository (2007 – present) is located at Vanderbilt University, Nashville, TN and was designed to link clinical data available from de-identified electronic medical records to DNA specimens.(Pulley et al., 2010) The BioVU Repository consists of de-identified blood samples obtained from patients at Vanderbilt University Medical Center Hospital, including all clinics that are part of the hospital system. De-identified data from multiple sources are available within BioVU, including diagnostic and procedure codes, basic demographics, discharge summaries, nursing notes, progress notes, health history, multi-disciplinary assessments, laboratory values, echocardiogram diagnoses, imaging reports, electronically derived data, and inpatient medication orders. All subjects (both UF cases and controls) selected from BioVU had diagnostic imaging with ultrasound, magnetic resonance imaging (MRI), or computed tomography (CT). Included as UF cases were women who had diagnostic imaging and either a diagnosis of a UF, as indicated by physician diagnosis of UFs or a surgical procedure for UF removal. For controls, two or more instances of pelvic imaging on separate dates were required. Initial chart review of a small subset of controls suggests that a large proportion of imagining information comes from prior pregnancy ultrasounds. Women with hysterectomy, myomectomy, or other procedures for UFs were excluded as controls. Controls were density matched to UF cases based on date of diagnostic imaging, where controls second imaging date had to be within a three to five year window of those cases. Both cases and controls were 18 to 65 years of age. We did not limit controls for age, but did perform secondary analyses limiting controls to those greater than 50 years of age to reduce the possibility that some women might develop a UF after imaging was performed. Our sampling algorithm to define UF cases and controls is informed by a published UF algorithm by Hartmann and colleagues using electronic medical records.(Hartmann et al., 2006) The IRB of Vanderbilt University, Nashville, TN approved this study.

SNP Selection

SNPs were selected based on either previously being associated with UFs in the GWAS by Cha and colleagues or being a haplotype-tagging SNP.(Cha et al., 2011) The SNPs with the strongest associations by Cha and colleagues were rs2280543 (chromosome 11, in the BET1L gene), rs12484776 (chromosome 22, in the TNRC6B gene), and rs7913069 (chromosome 10, not located within a gene, but referred to as “nearby SLK”). The remaining SNPs selected were haplotype-tagging SNPs near these loci. Haplotype-tagging SNPs were identified using the HapMap phase III samples (Release 28, http://www.hapmap.org): African American (from the ASW USA), Yoruban from Ibadan, Nigeria (YRI), and Northern and Western European (Centre d'Etude du Polymorphisme Humain (CEPH) family samples from Utah, USA), with the Tagster htSNP linkage disequilibrium (LD) selection tool available from the SNPinfo Web Server (National Institute for Environmental Health Sciences; http://snpinfo.niehs.nih.gov/). Within each reference population selected SNPs had a minor allele frequency (MAF) of 0.10 or greater, were in bins of highly correlated SNPs (r2 greater than or equal to 0.80), and were located within five kilobases from the boundaries of candidate genes and/or SNPs if the index SNP was not located within a gene. The GWAS index SNPs were forced into the Tagster htSNP selection algorithm and through an iterative approach, a minimum set of htSNPs for study subjects with admixed ancestry were identified.(Thorisson et al., 2005;Xu et al., 2007) A total of 72 SNPs met the above criteria. A summary of the SNPs used in our final analyses is provided in Table 1 and gene schematics are provided on Supplemental Figures 1 through 3. Information regarding SNP location within a gene, its type, and any corresponding amino acid changes (none were found) were sought from the HapMap and the SNPper program (http://snpper.chip.org/).

Table 1.

Final list of SNPs included in UF association analyses among the Right from the Start cohort (2001-2012)

rs# Location Type
BET1L gene Chromosome 11
+/- 5 kilobases 187,924-202,382
rs3741411 189,256 Intron
rs7114102 190,289 Downstream
rs939917 192,547 Downstream
rs11602954 192,856 Downstream
rs2280543* 193,788 3' UTR
rs2280545 194,147 3' UTR
rs1045454 194,228 3' UTR
rs4980319 194,986 3' UTR
rs3782123 195,198 3' UTR
rs7930823 196,767 Intron
rs2293168 201,482 Intron

TNRC6B gene Chromosome 22
+/- 5 kilobases 38,765,767-39,066,757
rs7291300 38,770,164 Promoter
rs9611257 38,785,324 Intron
rs6001738 38,789,557 Intron
rs5995802 38,791,365 Intron
rs6001741 38,794,150 Intron
rs11912610 38,796,157 Intron
rs6001743 38,797,954 Intron
rs5995810 38,807,376 Intron
rs7292838 38,809,394 Intron
rs9607685 38,809,757 Intron
rs6001762 38,825,419 Intron
rs11705409 38,826,602 Intron
rs9611265 38,828,439 Intron
rs12157468 38,830,259 Intron
rs9611266 38,830,798 Intron
rs11913462 38,834,510 Intron
rs9611267 38,835,950 Intron
rs17001651 38,841,126 Intron
rs5995814 38,842,688 Intron
rs12628757 38,847,003 Intron
rs6001783 38,854,137 Intron
rs2413611 38,857,804 Intron
rs8140112 38,863,076 Intron
rs2143177 38,865,677 Intron
rs17323619 38,868,663 Intron
rs11089974 38,873,554 Intron
rs9611286 38,914,908 Intron
rs12628783 38,916,015 Intron
rs8137189 38,929,482 Intron
rs138019 38,941,574 Intron
rs3091342 38,942,102 Intron
rs138022 38,942,982 Intron
rs6001848 38,966,745 Intron
rs5750913 38,970,231 Intron
rs3752513 38,971,926 Intron (boundary)
rs12484776* 38,982,819 Intron
rs12628051 38,984,222 Intron
rs739181 38,986,834 Intron
rs4821940 38,989,519 Intron
rs6001862 39,011,734 Intron (boundary)
rs713898 39,013,786 Intron
rs5995843 39,027,323 Intron (boundary)
rs139909 39,027,527 Intron
rs139910 39,033,834 Intron
rs4821942 39,048,046 Intron
rs139916 39,051,008 3' UTR
rs139921 39,056,708 3' UTR
rs470113 39,059,560 3' UTR
rs12484697 39,066,418 Downstream/Promoter

rs7913069* (nearby SLK) Chromosome 10
+/- 5 kilobases 105,699,390-105,709,390
rs7079220 105700137 -
rs2864004 105701838 -
rs11191875 105702778 -
rs7913069* 105704389 -
rs4244255 105709231 Promoter
*

Index SNPs

DNA Extraction and Genotyping

Genotyping BioVU

BioVU DNA samples were isolated from whole blood using the Autopure LS system (QIAGEN Inc., Valencia, CA). In BioVU we only genotyped the top three associated SNPs from the previously published GWAS (rs7913069, rs2280543, and rs12484776) and they were all genotyped using a TaqMan allelic discrimination assay.

RFTS Genotyping

DNA for RFTS saliva samples was extracted using Oragene DNA (Genotek Inc., Ontario, Canada) manufacturer recommended DNA extraction procedures. In the RFTS population, one tag SNP (rs6519215) was genotyped using a TaqMan allelic discrimination assay purchased from the ABI Assay on Demand or Assays by Design services (Life Technologies, Grand Island, NY) . The remaining 71 SNPs were genotyped using the Sequenom MassARRAY genotyping platform (Sequenom Inc., San Diego, CA). One SNP (rs5757906) assay failed. The final analytic dataset for RFTS contained 65 SNPs. All SNPs in BioVU and RFTS had genotyping call rates of 95% or better (mean call rates of 98%) and QC sample match rates of 100%. Six SNPs were dropped because of low MAF (< 0.01) in the genotyped dataset.

Statistical Analysis

Tests for deviations from Hardy Weinberg Equilibrium (HWE) were performed using PLINK statistical software.(Purcell et al., 2007) Statistical significance for these analyses was determined using p values from Fisher's exact tests. Pairwise LD was characterized using the standard summary statistic r2 from HaploView(Barrett et al., 2005) statistical software, where r2 is the correlation of SNPs in a population that takes into account differences in allele frequencies and is less sensitive to inflation due to small sample size. Haplotype blocks were assigned, using the D’ confidence interval algorithm created by Gabriel et al.(Gabriel et al., 2002) Descriptive statistics of demographic data were expressed as frequencies and proportions and compared between women with and without UFs (reference) using unadjusted logistic regression using STATA 11.0 statistical software (College Station, TX).

Single locus tests of association with UF risk were performed using logistic regression assuming an additive genotypic model (0 (homozygous major allele) versus 1 (heterozygous) versus 2 (homozygous minor allele)). Odds ratios (ORs) and confidence intervals (CI) were reported for SNPs from all statistical models. We reported results from both regression models unadjusted and adjusted for potential confounders: age (categorical) and BMI (categorical). Unadjusted models are presented in the manuscript for comparison with the previous results from Cha and colleagues; adjusted models can be found in Supplemental Table 1. PLINK statistical software was used to perform single locus tests of association.(Purcell et al., 2007)

Single locus association analyses in RFTS and BioVU were further analyzed together with fixed-effects meta-analyses using PLINK as well as METAL.(Purcell et al., 2007;Willer et al., 2010) We only considered the fixed effects results among EAs from RFTS and BioVU. Thereby, we sought out only those loci with consistent evidence between the two populations using this approach.

RESULTS

Right from the Start (RFTS)

Fourteen percent of women from RFTS had UFs (n=89). Age greater than or equal to 30 years was associated with increased risk for UFs (Table 2A). None of the SNPs 65 haplotype-tagging SNPs examined significantly deviated from HWE. In unadjusted analyses, five SNPs associated (p ≤ 0.05) with increased risk of UFs among EAs (Table 3). Among these associated SNPs, one was in the 10q24.33 chromosomal region (rs11191875, OR = 2.78, 95% CI 1.24 to 6.26, p = 0.014), one was in BET1L (rs939917, OR = 1.86, 95% CI 1.12 to 3.07, p = 0.016;) and three were in TNRC6B (rs11089974, OR = 1.46, 95% CI 1.01 to 2.10, p = 0.046; rs12484776, OR = 1.48, 95% CI 1.03 to 2.13, p = 0.035; rs4821942, OR = 1.52, 95% CI 1.06 to 2.18, p = 0.024). The set of SNPs that were associated with UFs also showed evidence for association after adjustment for age and BMI (Supplemental Table 1).

Table 2.

Demographic characteristics and their associations with UFs in the Right from the Start (2001-2012) and BioVU (2007-present) cohorts

A. Right from the Start Cohort
Characteristic n No UFs (n = 552) % UFs (n = 89) % OR* 95% CI
Lower Upper
Age
    Less than 25 58 10 2 1.00 Reference
    25 to 29 230 38 21 2.52 0.57 11.15
    30 to 34 239 37 42 5.12 1.20 21.94
    35-49 114 15 35 10.46 2.41 45.46
    Greater than or equal to 50 0 0 0 - - -
Body mass index
    Underweight (less than 20) 68 11 7 0.74 0.32 1.74
    Normal weight (20 to 24.9) 307 48 41 1.00 Reference
    Overweight (25 to 29.9) 152 24 21 1.04 0.59 1.83
    Obese (30 and above) 114 17 20 1.38 0.77 2.48
Study site
    North Carolina 195 28 54 2.20 1.39 3.47
    Tennessee 444 72 46 1.00 Reference
    Texas 2 <1 0 - - -
B.
Characteristic n No UFs (n = 997) % UFs (n = 997) % OR 95% CI
Lower Upper
Age
    Less than 25 18 1 <1 1.00 Reference
    25 to 29 62 5 1 0.59 0.16 2.22
    30 to 34 106 8 3 1.45 0.44 4.74
    35-49 508 21 31 5.35 1.73 16.47
    50-64 726 28 46 5.71 1.86 17.51
    Greater than or equal to 65 527 36 18 1.75 0.57 5.41
Body mass index
    Underweight (less than 20) 117 7 5 0.75 0.50 1.13
    Normal weight (20 to 24.9) 526 29 28 1.00 Reference
    Overweight (25 to 29.9) 556 29 31 1.08 0.85 1.36
    Obese (30 and above) 670 35 36 1.06 0.84 1.33

n = number, OR = odds ratio, CI = confidence interval

Table 3.

Summary of unadjusted index SNP and strongest single locus associations with UFs among the Right from the Start and BioVU cohorts

Population Gene rs # MA MAF
OR 95% CI
P
No UFs UF Lower Upper
RFTS EA1 Nearby SLK rs11191875 T 0.02 0.05 2.78 1.24 6.26 0.014
rs7913069* T 0.01 0.01 0.42 0.05 3.20 0.400

BET1L rs939917 T 0.34 0.50 1.86 1.12 3.07 0.016
rs2280543* T 0.04 0.02 0.55 0.20 1.56 0.262

TNRC6B rs11089974 T 0.20 0.26 1.46 1.01 2.10 0.046
rs12484776* G 0.20 0.27 1.48 1.03 2.13 0.035
rs4821942 A 0.20 0.28 1.52 1.06 2.18 0.024

BioVU EA
Nearby SLK rs7913069* T 0.01 0.02 1.18 0.71 1.97 0.527

BET1L rs2280543* T 0.05 0.04 0.68 0.51 0.92 0.013

TNRC6B rs12484776* G 0.20 0.23 1.17 1.00 1.36 0.050
1

European Americans are all non-Hispanic

MA = major allele, MAF = minor allele frequency, OR = odds ratio, CI = confidence interval

*

Index SNP from previous GWAS

Highlighted results indicate p ≤ 0.05

Further examination of the LD structure among the SNPs in TNRC6B that associated with UF risk showed evidence for high LD between rs11089974, rs12484776, and rs4821942—with r2 values between these SNPs ranging from 0.88 to 0.97 in cases and 0.87 to 0.93 in controls (Supplemental Figure 4). Based on the strong LD observed between these TNRC6B SNPs, we performed single SNP association analyses conditioning on rs12484776 (GWAS index SNP).These analyses showed that none of the SNPs in TNRC6B were statistically significant after adjusting for rs12484776 in regression models (results not shown). This would suggest that associations at other SNPs in TNRC6B were due to being in LD with rs12484776.

BioVU

BioVU participants were on average older than RFTS study participants (Table 2). BioVU genotyping data were only for the top three previously associated GWAS SNPs. Fifty percent of women included in these analyses from BioVU had UFs. Similar to women from RFTS, older age was associated with increased risk for UFs (Table 2B). Greater proportions of women from BioVU had higher BMIs or were older than women in RFTS; this reflects RFTS samples coming from a younger cohort while BioVU represents a clinical population.

None of the SNPs examined significantly deviated from HWE. Among the three index SNPs examined for association with UF risk, two showed evidence for association in unadjusted analyses (BET1L rs2280543 OR = 0.68, 95% CI 0.51 to 0.92, p = 0.013; TNRC6B rs12484776 OR = 1.17, 95% CI 1.00 to 1.36, p = 0.050) (Table 3). This evidence for association with UFs remained after adjusting for age and BMI (Supplemental Table 1).

We were not able to determine the age at which a study participant developed UFs or if they develop a UF after being screened. To address this in BioVU we performed secondary analyses limiting BioVU controls to women over 50 (data not shown). Risk estimates for UF were larger when limiting BioVU controls to women over 50 (BET1L rs2280543 OR = 0.71, 95% CI 0.51 to 0.99, p = 0.042; TNRC6B rs12484776 OR = 1.26, 95% CI 1.05 to 1.51, p = 0.011) suggesting that despite observing consistent associations at index SNPs, the younger subset of controls may have been contributing to phenotypic heterogeneity.

RFTS BioVU meta-analyses

Meta-analyses across RFTS and BioVU samples showed strong evidence of association at BET1L rs2280543 (meta OR = 0.67, SE = 0.15, Q = 0.70, I = 0, p = 6.90×10-3) and TNRC6B rs12484776 (meta OR = 1.21, SE = 0.07, Q = 0.24, I = 28.37, p = 8.70×10-3) (Table 4). Finally, in order to assess the consistency of effect sizes and association results with the prior GWAS of a Japanese population, we did a meta-analysis including all RFTS participants, BioVU participants, and the prior Japanese GWAS (Table 4). Statistical significance was stronger for all three SNPs compared to the level of significance in the prior GWAS. Little evidence of heterogeneity across the study populations was indicated for these SNPs, with Q's ranging from 0.21 to 0.92 and I = 0 to 36.01. The SNP with the strongest meta-association p value across all populations in RFTS, BioVU, and the prior GWAS of Japanese subjects was BET1L rs2280543 (OR = 0.66, SE = 0.07, Q = 0.92, I = 0, p = 3.89×10-9), which associated with p = 7.16×10-7 in the paper by Cha and colleagues.(Cha et al., 2011) This level of statistical significance exceeds the canonical genome-wide threshold for multiple testing, using a Bonferroni correction for multiple testing. It is of note, however, that in the prior GWAS they used the major allele as the risk allele for rs2280543. We used the minor allele as the risk allele in our analyses and our results are consistent for rs2280543 when modeled with the same risk allele.

Table 4.

Unadjusted SNP and UF associations from meta-analyses across Right from the Start, BioVU, and a previously published GWAS of a Japanese population

Meta-Analysis Populations Gene MA rs # OR SE Q I P
RFTS EA and BioVU Nearby SLK T rs7913069* 1.11 0.25 0.33 0 0.682
BET1L T rs2280543* 0.67 0.15 0.70 0 6.9×10-3
TNRC6B G rs12484776* 1.21 0.07 0.24 28.37 8.7×10-3
RFTS EA, BIOVU EA, and Prior Japanese GWAS Nearby SLK T rs7913069* 1.58 0.08 0.21 36.01 7.45×10-8
BET1L T rs2280543* 0.66 0.07 0.92 0 3.89×10-9
TNRC6B G rs12484776* 1.26 0.04 0.38 0 1.33×10-8

OR = odds ratio, SE =standard error, Q = p value for the Cochrane's Q statistic, I = I2 heterogeneity index (0-100)

*

Index SNP from previous GWAS. (30)

Highlighted results indicate p ≤ 0.05

DISCUSSION

This study is the first replication of the associations previously observed in BET1L and TNRC6B in two EA U.S. cohorts and is enhanced by pelvic imaging for cases and controls. We observed strong evidence of association across several markers in BET1L and TNRC6B including two of the previously associated GWAS index SNPs. The strongest evidence for association came from our EA subset; however, we were underpowered to detect associations across the other racial groups. The direction of the effect sizes across SNPs in the prior Japanese GWAS and our study were consistent with little evidence of heterogeneity in effect sizes across studies. The very low heterogeneity of effects at these loci between European and Asian populations further support a consistent effect on risk and suggest that this locus may be functional or in tight LD with the functional SNP. We did not replicate the association previously observed at rs7913069 within RFTS or BioVU; however, the SNP was significant when we meta-analyzed including the prior GWAS with a higher level of statistical significance than was previously reported (GWAS p = 7.9×10-8). We note, however, we were less powered to detect an association among EAs at this SNP because the MAF was 0.01 while among the Japanese population the MAF was between 0.07 and 0.11.(Cha et al., 2011)

We note that the associations at the SNPs identified by Cha and colleagues did not previously replicate in a cohort of African American women from the Black Women's Health Study (BWHS) and were not among the top associations reported in a recent association study using women of European ancestry (U.S. European Americans and Australians).(Eggert et al., 2012;Wise et al., 2012) Inconsistencies in association results between our study and these previously published studies may be due to the genetic ancestry of the study participants, as all of our participants were EAs from the U.S. while the prior two studies consisted of African Americans and combined subjects of European ancestry from the U.S. and Australia. It may be that these SNPs associate among EAs from the U.S. and Japanese populations but not other racial or geographic groups. Furthermore, the phenotype definition used to define cases and controls by these prior studies was based on self-report, while our studies required imagining confirmation of fibroid status.

The strongest evidence for association with UF came from BET1L and TNRC6B. Neither BET1L nor TNRC6B were previously associated with UF risk, except for the GWAS by Cha and colleagues. According to the NHGRI Catalog of Published GWAS (http://www.genome.gov/gwastudies/), the BET1L SNP rs2280543 has also been associated with intracranial aneurysm in another GWAS in a Japanese population.(Low et al., 2012) Although rs12484776 has not been identified by other GWAS, other SNPs within the TNRC6B have been associated with both prostate cancer risk among EA and height.(Estrada et al., 2009;Liu et al., 2011;Sun et al., 2009;Tao et al., 2012) TNRC6B has been shown to interact with insulin-like growth factors 2 (IGF-2) to increase risk for prostate cancer.(Tao et al., 2012) Furthermore, quantitative trait loci within the region of TNRC6B have been shown to be associated with age-at-menarche and early age-at-menarche is an established risk factor for UF.(Dragomir et al., 2010;Faerstein et al., 2001;Guo et al., 2006;Lumbiganon et al., 1996;Marshall et al., 1998;Samadi et al., 1996;Wise et al., 2004) BET1L is involved in endoplasmic reticulum to golgi transport while TNRC6B is involved in RNA interference machinery and is important for miRNA RNA-dependent translational regression or degradation of target RNAs. TNRC6B is a potential biological target as miRNAs have previously implicated in leiomyoma pathogenesis.(Luo and Chegini, 2008;Meister et al., 2005)

Further examination of the genes near the GWAS index SNPs show strong evidence of those genes being involved in cardiovascular-related health conditions. OBFC1 has been associated with cardiovascular disease and SIRT3 with metabolic syndrome, mitochondrial function, obesity, and exercise response in prior studies.(Borengasser et al., 2011;Burnett-Hartman et al., 2012;Capel et al., 2008;Choudhury et al., 2011;Giralt and Villarroya, 2012;Green and Hirschey, 2012;Guarente, 2011;Mestre-Alfaro et al., 2012;Valdecantos et al., 2012;Vasan et al., 2007) Insertion/deletions within the BET1L chromosomal region have also been implicated in glucose regulation and type II diabetes.(Owerbach et al., 1982;Rotwein et al., 1981) These data suggest that genes associated with metabolic complications and cancer may also be involved with UF pathogenesis, which is interesting as being overweight is a risk factor for UFs.(Baird et al., 2007;Takeda et al., 2008;Terry et al., 2007;Wise et al., 2005) Further research is necessary to assess the possible role of genetic interactions with cardiovascular outcomes in UF risk.

There are no other established genetic risk factors for UFs. In addition to the recently published GWAS by Cha and colleagues(Cha et al., 2011) there have been three other prominently published large-scale genetic association studies.(Eggert et al., 2012;Makinen et al., 2011;Wise et al., 2012) These include a tumor sequencing study published by Mäkinen and colleagues published in the journal Science,(Makinen et al., 2011) a GWAS of UF using a EA family and population-based sample,(Eggert et al., 2012) and an admixture mapping analysis using a African American populations.(Wise et al., 2012) Among these only the study by Mäkinen and colleagues has been validated in multiple independent studies. The Eggert study observed one locus at genome-wide significance, but without an independent replication. Mäkinen and colleagues examined somatic mutations in tumor tissue and found most UFs had mutations at the gene mediator complex subunit 12 (MED12), a result that has replicated across independent multi-ethnic populations.(Je et al., 2012;Makinen et al., 2011) MED12 is a 26-subunit transcriptional regulator that bridges DNA regulatory sequences to the RNA polymerase II initiation complex. All associated mutations resided in exon 2, suggesting that aberrant function of this region of MED12 contributes to tumorigenesis. Although some recent research suggests that mutations in MED12 are specific to UF tissue,(Je et al., 2012) other studies suggest that MED12 may be involved in multiple pathways that contribute to tumor growth in other tissues.(Markowski et al., 2012) Further supporting the later hypothesis is a recent study published in Nature Genetics showing that MED12 mutations are also present in prostate cancer tumor tissue.(Barbieri et al., 2012) Further research can elucidate any relationship MED12 may have with the genes identified by Cha and colleagues.(30)

A significant strength of our study is that all women were systematically screened for UFs using a standardized protocol and endovaginal ultrasounds for RFTS and various forms of pelvic imaging for BioVU. The majority of other UF studies did not have imaging data available for all subjects, but instead relied on clinical diagnosis of UFs. As a result, misclassification of UFs within our cohorts should be very low. Additionally, although BioVU participants had a higher mean age than RFTS participants who were primarily in their 20s. It may be that women with UFs in the RFTS cohort represent a group with an early onset of the condition because estimates of age-specific cumulative incidence suggest that many women develop UFs later in their reproductive years.(Laughlin et al., 2010b)

Little is known about UF pathophysiology or genetic risk factors beyond what has been learned from cell culture studies and tumor biology. The GWAS by Cha and colleagues and our findings support that common germline variation may contribute to increased UF risk. When meta-analyzed across all cohorts, including the prior GWAS, the level of statistical significance across all three previously associated GWAS SNPs exceeds the canonical genome-wide threshold for multiple testing. Taken together these data support a consistent effect on risk and suggest that this locus may be functional or in tight LD with the functional SNP. Barriers often faced by UF researchers today include lack of imaging, limited racial diversity in cohorts, and availability of DNA samples. Our study population is unique, as all women included in this replication study had pelvic imaging available to confirm the presence or absence of a UF. Even though only a small number of genetic epidemiology studies have been performed, they have each yielded some important insights into the genetics of UF. Our findings suggest that there is common germline variation that increase risk for UFs among both EA and Japanese; however, further research is necessary in order to assess the role of BET1L and TNRC6B in other minority groups.

Supplementary Material

439_2013_1306_MOESM1_ESM

Supplemental Figure 1. Candidate and haplotype tagging SNPs near BET1L. BET1L is oriented 5’ to 3’ and labeled in center. In bold are labeled GWAS index SNPs.

439_2013_1306_MOESM2_ESM

Supplemental Figure 2. Candidate and haplotype tagging SNPs near SLK. SLK is oriented 5’ to 3’. In bold are labeled GWAS index SNPs.

439_2013_1306_MOESM3_ESM

Supplemental Figure 3. Candidate and haplotype tagging SNPs near TNRC6B. TNRC6B is oriented 5’ to 3’ and labeled in center. In bold are labeled GWAS index SNPs.

439_2013_1306_MOESM4_ESM

Supplemental Figure 4. Right from the Start linkage disequilibrium structure among European Americans. LD plots are presented for EAs cases and controls. All figures are oriented 5’ to 3’, right to left, relative to the minus strand. r2 (shades of black) are indicated in percentages within squares in the LD plots, with solid blocks with numbers indicating r2 = 1. Strong LD is indicated by dark gray and like gray and white indicate weak LD and/or low confidence values.

439_2013_1306_MOESM5_ESM

Supplemental Table 1. Summary of adjusted index SNP and strongest (p ≤ 0.05) single locus associations with UFs among the Right from the Start and BioVU cohorts

ACKNOWLEDGEMENTS

The field research for Right from the Start was supported by grants from the National Institute of Child and Human Development (R01HD043883 and R01HD049675) and the American Water Works Association Research Foundation (2579). Additional funds were provided by the Building Interdisciplinary Research Careers in Women's Health career development program (K12HD4383), the Vanderbilt Clinical and Translational Research Scholar Award 5KL2RR024975 to TLE, the Vanderbilt CTSA grant UL1 RR024975-01 from NCRR/NIH, and the BioVU dataset used for the analyses described was obtained from Vanderbilt University Medical Center's BioVU which is supported by institutional funding and by the Vanderbilt CTSA grant 1UL1RR024975-01 from NCRR/NIH.

Reference List

  1. Baird DD, Dunson DB. Why is parity protective for uterine fibroids? Epidemiology. 2003;14:247–250. doi: 10.1097/01.EDE.0000054360.61254.27. [DOI] [PubMed] [Google Scholar]
  2. Baird DD, Dunson DB, Hill MC, Cousins D, Schectman JM. High cumulative incidence of uterine leiomyoma in black and white women: ultrasound evidence. Am J Obstet Gynecol. 2003;188:100–107. doi: 10.1067/mob.2003.99. [DOI] [PubMed] [Google Scholar]
  3. Baird DD, Dunson DB, Hill MC, Cousins D, Schectman JM. Association of physical activity with development of uterine leiomyoma. Am J Epidemiol. 2007;165:157–163. doi: 10.1093/aje/kwj363. [DOI] [PubMed] [Google Scholar]
  4. Barbieri CE, Baca SC, Lawrence MS, Demichelis F, Blattner M, Theurillat JP, White TA, Stojanov P, Van AE, Stransky N, Nickerson E, Chae SS, Boysen G, Auclair D, Onofrio RC, Park K, Kitabayashi N, MacDonald TY, Sheikh K, Vuong T, Guiducci C, Cibulskis K, Sivachenko A, Carter SL, Saksena G, Voet D, Hussain WM, Ramos AH, Winckler W, Redman MC, Ardlie K, Tewari AK, Mosquera JM, Rupp N, Wild PJ, Moch H, Morrissey C, Nelson PS, Kantoff PW, Gabriel SB, Golub TR, Meyerson M, Lander ES, Getz G, Rubin MA, Garraway LA. Exome sequencing identifies recurrent SPOP, FOXA1 and MED12 mutations in prostate cancer. Nat Genet. 2012;44:685–689. doi: 10.1038/ng.2279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Barrett JC, Fry B, Maller J, Daly MJ. Haploview: analysis and visualization of LD and haplotype maps. Bioinformatics. 2005;21:263–265. doi: 10.1093/bioinformatics/bth457. [DOI] [PubMed] [Google Scholar]
  6. Bonatz G, Frahm SO, Andreas S, Heidorn K, Jonat W, Parwaresch R. Telomere shortening in uterine leiomyomas. Am J Obstet Gynecol. 1998;179:591–596. doi: 10.1016/s0002-9378(98)70050-x. [DOI] [PubMed] [Google Scholar]
  7. Borengasser SJ, Lau F, Kang P, Blackburn ML, Ronis MJ, Badger TM, Shankar K. Maternal obesity during gestation impairs fatty acid oxidation and mitochondrial SIRT3 expression in rat offspring at weaning. PLoS One. 2011;6:e24068. doi: 10.1371/journal.pone.0024068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Burnett-Hartman AN, Fitzpatrick AL, Kronmal RA, Psaty BM, Jenny NS, Bis JC, Tracy RP, Kimura M, Aviv A. Telomere-associated polymorphisms correlate with cardiovascular disease mortality in Caucasian women: The Cardiovascular Health Study. Mech Ageing Dev. 2012;133:275–281. doi: 10.1016/j.mad.2012.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Capel F, Viguerie N, Vega N, Dejean S, Arner P, Klimcakova E, Martinez JA, Saris WH, Holst C, Taylor M, Oppert JM, Sorensen TI, Clement K, Vidal H, Langin D. Contribution of energy restriction and macronutrient composition to changes in adipose tissue gene expression during dietary weight-loss programs in obese women. J Clin Endocrinol Metab. 2008;93:4315–4322. doi: 10.1210/jc.2008-0814. [DOI] [PubMed] [Google Scholar]
  10. Cha PC, Takahashi A, Hosono N, Low SK, Kamatani N, Kubo M, Nakamura Y. A genome-wide association study identifies three loci associated with susceptibility to uterine fibroids. Nat Genet. 2011;43:447–450. doi: 10.1038/ng.805. [DOI] [PubMed] [Google Scholar]
  11. Choudhury M, Jonscher KR, Friedman JE. Reduced mitochondrial function in obesity-associated fatty liver: SIRT3 takes on the fat. Aging (Albany NY) 2011;3:175–178. doi: 10.18632/aging.100289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Cramer SF, Patel A. The frequency of uterine leiomyomas. Am J Clin Pathol. 1990;94:435–438. doi: 10.1093/ajcp/94.4.435. [DOI] [PubMed] [Google Scholar]
  13. Dragomir AD, Schroeder JC, Connolly A, Kupper LL, Hill MC, Olshan AF, Baird DD. Potential risk factors associated with subtypes of uterine leiomyomata. Reprod Sci. 2010;17:1029–1035. doi: 10.1177/1933719110376979. [DOI] [PubMed] [Google Scholar]
  14. Eggert SL, Huyck KL, Somasundaram P, Kavalla R, Stewart EA, Lu AT, Painter JN, Montgomery GW, Medland SE, Nyholt DR, Treloar SA, Zondervan KT, Heath AC, Madden PA, Rose L, Buring JE, Ridker PM, Chasman DI, Martin NG, Cantor RM, Morton CC. Genome-wide linkage and association analyses implicate FASN in predisposition to Uterine Leiomyomata. Am J Hum Genet. 2012;91:621–628. doi: 10.1016/j.ajhg.2012.08.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. El-Gharib MN, Elsobky ES. Cytogenetic aberrations and the development of uterine leiomyomata. J Obstet Gynaecol Res. 2010;36:101–107. doi: 10.1111/j.1447-0756.2009.01099.x. [DOI] [PubMed] [Google Scholar]
  16. Estrada K, Krawczak M, Schreiber S, van DK, Stolk L, van Meurs JB, Liu F, Penninx BW, Smit JH, Vogelzangs N, Hottenga JJ, Willemsen G, de Geus EJ, Lorentzon M, von Eller-Eberstein H, Lips P, Schoor N, Pop V, de KJ, Hofman A, Aulchenko YS, Oostra BA, Ohlsson C, Boomsma DI, Uitterlinden AG, van Duijn CM, Rivadeneira F, Kayser M. A genome-wide association study of northwestern Europeans involves the C-type natriuretic peptide signaling pathway in the etiology of human height variation. Hum Mol Genet. 2009;18:3516–3524. doi: 10.1093/hmg/ddp296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Faerstein E, Szklo M, Rosenshein NB. Risk factors for uterine leiomyoma: a practice-based case-control study. II. Atherogenic risk factors and potential sources of uterine irritation. Am J Epidemiol. 2001;153:11–19. doi: 10.1093/aje/153.1.11. [DOI] [PubMed] [Google Scholar]
  18. Flake GP, Andersen J, Dixon D. Etiology and pathogenesis of uterine leiomyomas: a review. Environ Health Perspect. 2003;111:1037–1054. doi: 10.1289/ehp.5787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Gabriel SB, Schaffner SF, Nguyen H, Moore JM, Roy J, Blumenstiel B, Higgins J, DeFelice M, Lochner A, Faggart M, Liu-Cordero SN, Rotimi C, Adeyemo A, Cooper R, Ward R, Lander ES, Daly MJ, Altshuler D. The structure of haplotype blocks in the human genome. Science. 2002;296:2225–2229. doi: 10.1126/science.1069424. [DOI] [PubMed] [Google Scholar]
  20. Giralt A, Villarroya F. SIRT3, a pivotal actor in mitochondrial functions: metabolism, cell death and aging. Biochem J. 2012;444:1–10. doi: 10.1042/BJ20120030. [DOI] [PubMed] [Google Scholar]
  21. Green MF, Hirschey MD. SIRT3 Weighs Heavily in the Metabolic Balance: A New Role for SIRT3 in Metabolic Syndrome. J Gerontol A Biol Sci Med Sci. 2012 doi: 10.1093/gerona/gls132. [DOI] [PubMed] [Google Scholar]
  22. Guarente L. Sirtuins, Aging, and Metabolism. Cold Spring Harb Symp Quant Biol. 2011 doi: 10.1101/sqb.2011.76.010629. [DOI] [PubMed] [Google Scholar]
  23. Guo Y, Shen H, Xiao P, Xiong DH, Yang TL, Guo YF, Long JR, Recker RR, Deng HW. Genomewide linkage scan for quantitative trait loci underlying variation in age at menarche. J Clin Endocrinol Metab. 2006;91:1009–1014. doi: 10.1210/jc.2005-2179. [DOI] [PubMed] [Google Scholar]
  24. Hartmann KE, Birnbaum H, Ben-Hamadi R, Wu EQ, Farrell MH, Spalding J, Stang P. Annual costs associated with diagnosis of uterine leiomyomata. Obstet Gynecol. 2006;108:930–937. doi: 10.1097/01.AOG.0000234651.41000.58. [DOI] [PubMed] [Google Scholar]
  25. Je EM, Kim MR, Min KO, Yoo NJ, Lee SH. Mutational analysis of MED12 exon 2 in uterine leiomyoma and other common tumors. Int J Cancer. 2012;131:E1044–E1047. doi: 10.1002/ijc.27610. [DOI] [PubMed] [Google Scholar]
  26. Kurbanova MK, Koroleva AG, Sergeev AS. [Genetic-epidemiologic analysis of uterine myoma: assessment of repeated risk]. Genetika. 1989;25:1896–1898. [PubMed] [Google Scholar]
  27. Laughlin SK, Baird DD, Savitz DA, Herring AH, Hartmann KE. Prevalence of uterine leiomyomas in the first trimester of pregnancy: an ultrasound-screening study. Obstet Gynecol. 2009;113:630–635. doi: 10.1097/AOG.0b013e318197bbaf. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Laughlin SK, Hartmann KE, Baird DD. Postpartum factors and natural fibroid regression. Am J Obstet Gynecol. 2011;204:496. doi: 10.1016/j.ajog.2011.02.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Laughlin SK, Herring AH, Savitz DA, Olshan AF, Fielding JR, Hartmann KE, Baird DD. Pregnancy-related fibroid reduction. Fertil Steril. 2010a;94:2421–2423. doi: 10.1016/j.fertnstert.2010.03.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Laughlin SK, Schroeder JC, Baird DD. New directions in the epidemiology of uterine fibroids. Semin Reprod Med. 2010b;28:204–217. doi: 10.1055/s-0030-1251477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Liu H, Wang B, Han C. Meta-analysis of genome-wide and replication association studies on prostate cancer. Prostate. 2011;71:209–224. doi: 10.1002/pros.21235. [DOI] [PubMed] [Google Scholar]
  32. Low SK, Takahashi A, Cha PC, Zembutsu H, Kamatani N, Kubo M, Nakamura Y. Genome-wide association study for intracranial aneurysm in the Japanese population identifies three candidate susceptible loci and a functional genetic variant at EDNRA. Hum Mol Genet. 2012;21:2102–2110. doi: 10.1093/hmg/dds020. [DOI] [PubMed] [Google Scholar]
  33. Lumbiganon P, Rugpao S, Phandhu-fung S, Laopaiboon M, Vudhikamraksa N, Werawatakul Y. Protective effect of depot-medroxyprogesterone acetate on surgically treated uterine leiomyomas: a multicentre case--control study. Br J Obstet Gynaecol. 1996;103:909–914. doi: 10.1111/j.1471-0528.1996.tb09911.x. [DOI] [PubMed] [Google Scholar]
  34. Luo X, Chegini N. The expression and potential regulatory function of microRNAs in the pathogenesis of leiomyoma. Semin Reprod Med. 2008;26:500–514. doi: 10.1055/s-0028-1096130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Luoto R, Kaprio J, Rutanen EM, Taipale P, Perola M, Koskenvuo M. Heritability and risk factors of uterine fibroids--the Finnish Twin Cohort study. Maturitas. 2000;37:15–26. doi: 10.1016/s0378-5122(00)00160-2. [DOI] [PubMed] [Google Scholar]
  36. Makinen N, Heinonen HR, Moore S, Tomlinson IP, van der Spuy ZM, Aaltonen LA. MED12 exon 2 mutations are common in uterine leiomyomas from South African patients. Oncotarget. 2011;2:966–969. doi: 10.18632/oncotarget.370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Malik M, Norian J, McCarthy-Keith D, Britten J, Catherino WH. Why leiomyomas are called fibroids: the central role of extracellular matrix in symptomatic women. Semin Reprod Med. 2010;28:169–179. doi: 10.1055/s-0030-1251475. [DOI] [PubMed] [Google Scholar]
  38. Markowski DN, Bartnitzke S, Loning T, Drieschner N, Helmke BM, Bullerdiek J. MED12 mutations in uterine fibroids-their relationship to cytogenetic subgroups. Int J Cancer. 2012;131:1528–1536. doi: 10.1002/ijc.27424. [DOI] [PubMed] [Google Scholar]
  39. Marsh EE, Lin Z, Yin P, Milad M, Chakravarti D, Bulun SE. Differential expression of microRNA species in human uterine leiomyoma versus normal myometrium. Fertil Steril. 2008;89:1771–1776. doi: 10.1016/j.fertnstert.2007.05.074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Marshall LM, Spiegelman D, Barbieri RL, Goldman MB, Manson JE, Colditz GA, Willett WC, Hunter DJ. Variation in the incidence of uterine leiomyoma among premenopausal women by age and race. Obstet Gynecol. 1997;90:967–973. doi: 10.1016/s0029-7844(97)00534-6. [DOI] [PubMed] [Google Scholar]
  41. Marshall LM, Spiegelman D, Goldman MB, Manson JE, Colditz GA, Barbieri RL, Stampfer MJ, Hunter DJ. A prospective study of reproductive factors and oral contraceptive use in relation to the risk of uterine leiomyomata. Fertil Steril. 1998;70:432–439. doi: 10.1016/s0015-0282(98)00208-8. [DOI] [PubMed] [Google Scholar]
  42. Meister G, Landthaler M, Peters L, Chen PY, Urlaub H, Luhrmann R, Tuschl T. Identification of novel argonaute-associated proteins. Curr Biol. 2005;15:2149–2155. doi: 10.1016/j.cub.2005.10.048. [DOI] [PubMed] [Google Scholar]
  43. Mestre-Alfaro A, Ferrer MD, Banquells M, Riera J, Drobnic F, Sureda A, Tur JA, Pons A. Body temperature modulates the antioxidant and acute immune responses to exercise. Free Radic Res. 2012;46:799–808. doi: 10.3109/10715762.2012.680193. [DOI] [PubMed] [Google Scholar]
  44. Moore AB, Flake GP, Swartz CD, Heartwell G, Cousins D, Haseman JK, Kissling GE, Sidawy MK, Dixon D. Association of race, age and body mass index with gross pathology of uterine fibroids. J Reprod Med. 2008;53:90–96. [PubMed] [Google Scholar]
  45. Myers SL, Baird DD, Olshan AF, Herring AH, Schroeder JC, Nylander-French LA, Hartmann KE. Self-report versus ultrasound measurement of uterine fibroid status. J Womens Health (Larchmt ) 2012;21:285–293. doi: 10.1089/jwh.2011.3008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Ojeda VJ. The pathology of hysterectomy specimens. N Z Med J. 1979;89:169–171. [PubMed] [Google Scholar]
  47. Owerbach D, Johansen K, Billesbolle P, Poulsen S, Schroll M, Nerup J. Possible association between DNA sequences flanking the insulin gene and atherosclerosis. Lancet. 1982;2:1291–1293. doi: 10.1016/s0140-6736(82)91505-7. [DOI] [PubMed] [Google Scholar]
  48. Parazzini F, Negri E, La VC, Chatenoud L, Ricci E, Guarnerio P. Reproductive factors and risk of uterine fibroids. Epidemiology. 1996;7:440–442. doi: 10.1097/00001648-199607000-00018. [DOI] [PubMed] [Google Scholar]
  49. Promislow JH, Makarushka CM, Gorman JR, Howards PP, Savitz DA, Hartmann KE. Recruitment for a community-based study of early pregnancy: the Right From The Start study. Paediatr Perinat Epidemiol. 2004;18:143–152. doi: 10.1111/j.1365-3016.2003.00546.x. [DOI] [PubMed] [Google Scholar]
  50. Pulley J, Clayton E, Bernard GR, Roden DM, Masys DR. Principles of human subjects protections applied in an opt-out, de-identified biobank. Clin Transl Sci. 2010;3:42–48. doi: 10.1111/j.1752-8062.2010.00175.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Purcell S, Neale B, Todd-Brown K, Thomas L, Ferreira MA, Bender D, Maller J, Sklar P, de Bakker PI, Daly MJ, Sham PC. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am J Hum Genet. 2007;81:559–575. doi: 10.1086/519795. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Rotwein P, Chyn R, Chirgwin J, Cordell B, Goodman HM, Permut MA. Polymorphism in the 5′-flanking region of the human insulin gene and its possible relation to type 2 diabetes. Science. 1981;213:1117–1120. doi: 10.1126/science.6267694. [DOI] [PubMed] [Google Scholar]
  53. Samadi AR, Lee NC, Flanders WD, Boring JR, III, Parris EB. Risk factors for self-reported uterine fibroids: a case-control study. Am J Public Health. 1996;86:858–862. doi: 10.2105/ajph.86.6.858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Snieder H, MacGregor AJ, Spector TD. Genes control the cessation of a woman's reproductive life: a twin study of hysterectomy and age at menopause. J Clin Endocrinol Metab. 1998;83:1875–1880. doi: 10.1210/jcem.83.6.4890. [DOI] [PubMed] [Google Scholar]
  55. Sozen I, Arici A. Interactions of cytokines, growth factors, and the extracellular matrix in the cellular biology of uterine leiomyomata. Fertil Steril. 2002;78:1–12. doi: 10.1016/s0015-0282(02)03154-0. [DOI] [PubMed] [Google Scholar]
  56. Sun J, Zheng SL, Wiklund F, Isaacs SD, Li G, Wiley KE, Kim ST, Zhu Y, Zhang Z, Hsu FC, Turner AR, Stattin P, Liu W, Kim JW, Duggan D, Carpten J, Isaacs W, Gronberg H, Xu J, Chang BL. Sequence variants at 22q13 are associated with prostate cancer risk. Cancer Res. 2009;69:10–15. doi: 10.1158/0008-5472.CAN-08-3464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Takeda T, Sakata M, Isobe A, Miyake A, Nishimoto F, Ota Y, Kamiura S, Kimura T. Relationship between metabolic syndrome and uterine leiomyomas: a case-control study. Gynecol Obstet Invest. 2008;66:14–17. doi: 10.1159/000114250. [DOI] [PubMed] [Google Scholar]
  58. Tao S, Wang Z, Feng J, Hsu FC, Jin G, Kim ST, Zhang Z, Gronberg H, Zheng LS, Isaacs WB, Xu J, Sun J. A genome-wide search for loci interacting with known prostate cancer risk-associated genetic variants. Carcinogenesis. 2012;33:598–603. doi: 10.1093/carcin/bgr316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Terry KL, De V,I, Hankinson SE, Spiegelman D, Wise LA, Missmer SA. Anthropometric characteristics and risk of uterine leiomyoma. Epidemiology. 2007;18:758–763. doi: 10.1097/EDE.0b013e3181567eed. [DOI] [PubMed] [Google Scholar]
  60. Thorisson GA, Smith AV, Krishnan L, Stein LD. The International HapMap Project Web site. Genome Res. 2005;15:1592–1593. doi: 10.1101/gr.4413105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Treloar SA, Martin NG, Dennerstein L, Raphael B, Heath AC. Pathways to hysterectomy: insights from longitudinal twin research. Am J Obstet Gynecol. 1992;167:82–88. doi: 10.1016/s0002-9378(11)91632-9. [DOI] [PubMed] [Google Scholar]
  62. Valdecantos MP, Perez-Matute P, Gonzalez-Muniesa P, Prieto-Hontoria PL, Moreno-Aliaga MJ, Martinez JA. Lipoic Acid Improves Mitochondrial Function in Nonalcoholic Steatosis Through the Stimulation of Sirtuin 1 and Sirtuin 3. Obesity (Silver Spring) 2012 doi: 10.1038/oby.2012.32. [DOI] [PubMed] [Google Scholar]
  63. Vasan RS, Larson MG, Aragam J, Wang TJ, Mitchell GF, Kathiresan S, Newton-Cheh C, Vita JA, Keyes MJ, O'Donnell CJ, Levy D, Benjamin EJ. Genome-wide association of echocardiographic dimensions, brachial artery endothelial function and treadmill exercise responses in the Framingham Heart Study. BMC Med Genet. 2007;8(Suppl 1):S2. doi: 10.1186/1471-2350-8-S1-S2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Vollenhoven B. Introduction: the epidemiology of uterine leiomyomas. Baillieres Clin Obstet Gynaecol. 1998;12:169–176. doi: 10.1016/s0950-3552(98)80059-x. [DOI] [PubMed] [Google Scholar]
  65. Willer CJ, Li Y, Abecasis GR. METAL: fast and efficient meta-analysis of genomewide association scans. Bioinformatics. 2010;26:2190–2191. doi: 10.1093/bioinformatics/btq340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Wise LA, Palmer JR, Harlow BL, Spiegelman D, Stewart EA, Adams-Campbell LL, Rosenberg L. Reproductive factors, hormonal contraception, and risk of uterine leiomyomata in African-American women: a prospective study. Am J Epidemiol. 2004;159:113–123. doi: 10.1093/aje/kwh016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Wise LA, Palmer JR, Spiegelman D, Harlow BL, Stewart EA, Adams-Campbell LL, Rosenberg L. Influence of body size and body fat distribution on risk of uterine leiomyomata in U.S. black women. Epidemiology. 2005;16:346–354. doi: 10.1097/01.ede.0000158742.11877.99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Wise LA, Ruiz-Narvaez EA, Palmer JR, Cozier YC, Tandon A, Patterson N, Radin RG, Rosenberg L, Reich D. African ancestry and genetic risk for uterine leiomyomata. Am J Epidemiol. 2012;176:1159–1168. doi: 10.1093/aje/kws276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Xu Z, Kaplan NL, Taylor JA. TAGster: efficient selection of LD tag SNPs in single or multiple populations. Bioinformatics. 2007;23:3254–3255. doi: 10.1093/bioinformatics/btm426. [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

439_2013_1306_MOESM1_ESM

Supplemental Figure 1. Candidate and haplotype tagging SNPs near BET1L. BET1L is oriented 5’ to 3’ and labeled in center. In bold are labeled GWAS index SNPs.

439_2013_1306_MOESM2_ESM

Supplemental Figure 2. Candidate and haplotype tagging SNPs near SLK. SLK is oriented 5’ to 3’. In bold are labeled GWAS index SNPs.

439_2013_1306_MOESM3_ESM

Supplemental Figure 3. Candidate and haplotype tagging SNPs near TNRC6B. TNRC6B is oriented 5’ to 3’ and labeled in center. In bold are labeled GWAS index SNPs.

439_2013_1306_MOESM4_ESM

Supplemental Figure 4. Right from the Start linkage disequilibrium structure among European Americans. LD plots are presented for EAs cases and controls. All figures are oriented 5’ to 3’, right to left, relative to the minus strand. r2 (shades of black) are indicated in percentages within squares in the LD plots, with solid blocks with numbers indicating r2 = 1. Strong LD is indicated by dark gray and like gray and white indicate weak LD and/or low confidence values.

439_2013_1306_MOESM5_ESM

Supplemental Table 1. Summary of adjusted index SNP and strongest (p ≤ 0.05) single locus associations with UFs among the Right from the Start and BioVU cohorts

RESOURCES