Next Article in Journal
Problem Formulation in Knowledge Discovery via Data Analytics (KDDA) for Environmental Risk Management
Previous Article in Journal
High Contributions of Secondary Inorganic Aerosols to PM2.5 under Polluted Levels at a Regional Station in Northern China
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Increased Urinary Phthalate Levels in Women with Uterine Leiomyoma: A Case-Control Study

1
Department of Obstetrics and Gynecology, Ilsan Paik Hospital, Inje University, 170 Joohwa-ro, Goyang-si, Kyeogi-do 10380, Korea
2
Department of Health, Environment and Safety, School of Human & Environmental Service, Eulji University, 553 Sansung-daero, Seongnam-si, Kyeogi-do 13135, Korea
3
Department of Biomedical and Pharmaceutical Sciences, College of Health Professions and Biomedical Sciences, University of Montana, 32 Campus Drive, Skaggs 283, Missoula, MT 59812, USA
*
Author to whom correspondence should be addressed.
Int. J. Environ. Res. Public Health 2016, 13(12), 1247; https://0-doi-org.brum.beds.ac.uk/10.3390/ijerph13121247
Submission received: 28 September 2016 / Revised: 9 December 2016 / Accepted: 11 December 2016 / Published: 15 December 2016
(This article belongs to the Section Environmental Health)

Abstract

:
We assessed the urinary concentration of 16 phthalate metabolites in 57 women with and without uterine leiomyoma (n = 30 and 27; respectively) to determine the association between phthalate exposure and uterine leiomyoma. To evaluate exposure to di-(2-ethylhexyl) phthalate (DEHP); we calculated the molar sum of DEHP metabolites; ∑3-DEHP (combining mono-(2-ethylhexyl) phthalate (MEHP); mono-(2-ethyl-5-hydroxyhexyl) phthalate (MEHHP); and mono-(2-ethyl-5-oxohexyl) phthalate); ∑4-DEHP (∑3-DEHP plus mono-(2-ethyl-5-carboxypentyl) phthalate); and ∑5-DEHP (∑4-DEHP plus mono (2-(carboxylmethyl)hexyl) phthalate (2cx-MMHP)). The log transformed urinary levels of MEHP; MEHHP; 2cx-MMHP; ∑3-DEHP; ∑4-DEHP; and ∑5-DEHP in the leiomyoma group were significantly higher than those of controls. When we adjusted for age; waist circumference; and parity using multiple logistic regression analyses; we found log ∑3-DEHP (OR = 10.82; 95% CI = 1.25; 93.46) and ∑4-DEHP (OR = 8.78; 95% CI = 1.03; 75.29) were significantly associated with uterine leiomyoma. Our findings suggest an association between phthalate exposure and uterine leiomyoma. However; larger studies are needed to investigate potential interactions between phthalate exposure and uterine leiomyoma.

1. Introduction

Myoma, adenomyoma, and endometriosis are estrogen-dependent diseases with unknown etiologies. Uterine leiomyomas are a type of myoma defined as benign tumors of uterine smooth muscles that represent a significant gynecologic disease resulting in reproductive dysfunction and pelvic pain. In particular, uterine leiomyoma is known to be a leading indication for hysterectomy [1,2], and its prevalence is relatively high, although it varies by ethnic group [3,4]. Baird et al. reported that the cumulative incidence of leiomyomas by age 50 in white women is 70% and exceeds 80% in black women in United States [5].
Phthalates are man-made chemicals that have been used in various industrial areas and are thought to be estrogenic and androgenic endocrine disruptors, although the evidence is controversial. Human exposure to phthalates is known to occur through inhalation in the workplace, exposure from automobile parts, exposure from building materials, and in the dermal application of personal care products and cosmetics as well as from children’s toys [6,7]. Additionally, exposure occurs through ingestion of water and food contaminated from food processing and packing materials and parenteral exposure through medical devices such as polyvinyl chloride (PVC) tubes and blood bags [8,9]. It has been shown that phthalates are high-volume production chemicals that are rapidly metabolized and excreted from the body [10].
Some phthalates, including di-(2-ethylhexyl) phthalate (DEHP) have been shown to alter or mimic estradiol in vivo and in vitro [11,12,13]. Phthalates prolong the menstrual cycle and increase the rate of premature menopause in animal models [14,15]. However, the adverse effects of phthalates on the human reproductive system are largely unknown. Many studies suggest that phthalates contribute to the pathogenesis of endometriosis [16,17,18]. Similar to endometriosis, leiomyoma are estrogen sensitive; therefore, endocrine-disrupting chemicals may play a role in their development. In fact, phthalates and diethylstilbestrol are reportedly associated with the prevalence of uterine leiomyoma [19,20]. However, there are very few studies examining the effects of phthalates on the development of uterine leiomyoma, and results have been controversial [21,22,23,24,25,26]. In particular, these studies have relied on self-reported fibroids [20,23] or small sample sizes [22,24,25]. The National Health and Nutrition Examination Survey (NHANES, 1999–2004) reported that mono-(2-ethylhexyl) phthalate (MEHP) is inversely associated with uterine leiomyoma, while monobutyl phthalate (MnBP) levels are positively associated with self-reported uterine leiomyoma [20]. Wise and colleagues also reported that the use of hair relaxer, a possible proxy for phthalate exposure, is associated with an increased incidence of self-reported uterine leiomyoma in a cohort of African-American women [23]. However, many women with leiomyomas are not aware of their disease; thus, self-reporting of uterine leiomyoma is a major limitation of these studies [27]. Furthermore, a small case-control study consisting of 36 leiomyoma cases and 29 controls showed that MEHP levels were higher in leiomyoma patients than in controls [24]. Another case-control study with 15 cases and 20 controls found lower serum levels of DEHP in leiomyoma, although the study participants did not present with disease symptoms and were limited to Caucasian women [25]. Recently, an operative cohort study joining 14 clinical centers and consisting of 99 cases and 374 controls reported no significant associations between the phthalates under investigation and odds of a leiomyoma diagnosis in either the unadjusted or adjusted analysis [26]; however, results from previous studies justify further examining the relationship between phthalate exposures and risk of uterine leiomyoma.
We hypothesized that exposures to DEHP metabolites (including MEHP) and MnBP are associated with risk of uterine leiomyoma. In the present study, we assessed urinary concentrations of 16 phthalate metabolites between women with and without uterine leiomyoma to explore the possible association between phthalate exposure and leiomyoma.

2. Materials and Methods

2.1. Clinical Subjects

We recruited patients who had undergone laparoscopic surgery and exploratory laparotomy between March 2013 and July 2015 at the department of Obstetrics & Gynecology in Inje University Ilsan Paik Hospital. Written informed consents were obtained from each patient using consent forms and a protocol approved by the Institutional Review Board of Ilsan Paik Hospital (IB-1211-039). All study participants were initially examined by gynecologic ultrasonography to detect uterine leiomyoma and any other gynecologic problems including intramural, submucosal, and subserosal fibroids before operation. Final uterine leiomyoma diagnoses were confirmed by direct visualization during operation and subsequent pathological analysis. Women with a history of occupational exposure to reproductive toxicants, hormone therapy, any malignancy or other reproductive treatment were excluded from this study. The indication for surgery in the control cases (n = 27) was benign ovarian cyst (n = 22) and carcinoma in situ (CIS) of the uterine cervix (n = 5).
Initially, the leiomyoma patient group consisted of 49 cases; we excluded cases with pathological evidence of endometriosis (n = 9), adenomyosis (n = 7), and both endometriosis and adenomyosis (n = 3) based on the findings from Huang et al., which showed a significant association between those conditions and an increased level of urinary phthalate metabolites [27]. Therefore, 30 cases of uterine leiomyoma were included in this study.

2.2. Demographic Characteristics

The demographic data of cases were obtained from an interviewed questionnaire before operation. The questions included age, body mass index (BMI), waist circumference, age of menarche, duration of menstrual cycle, history of dysmenorrhea and operation, gravity, parity, cigarette smoking, alcohol consumption, exposure to second hand smoke, and exercise.

2.3. Urine Collection and Analysis of Urinary Phthalate Metabolites

Gynecologic ultrasonography was used to confirm the presence of fibroids and the need for surgery. Once confirmed, all subjects were requested to undergo blood tests in preparation for surgery and before hospitalization. Urine samples (20–30 mL) were collected in phthalate-free polypropylene containers at the time of blood tests; surgery was scheduled 1–3 days later. Urine samples were immediately centrifuged and stored at −80 °C until further analysis and after measurement of creatinine levels.
Sixteen urinary phthalate metabolites were analyzed with liquid chromatography electrospray ionization mass spectrometry (LC-MS/MS) using Nanospace SI-2 (Shiseido, Tokyo, Japan) and API 4000 (Applied Biosystems, Foster, CA, USA) as detailed in a previous publication [28]. Standards of phthalate metabolites, some of the compounds listed are not metabolites but original phthalates including MEHP, MnBP, mono-iso-butyl phthalate (MiBP), mono-benzyl phthalate (MBzP), mono-cyclohexyl phthalate (MCHP), mono-(2-ethyl-5-hydroxyhexyl) phthalate (MEHHP), mono-(2-ethyl-5-oxohexyl) phthalate (MEOHP), mono-(2-ethyl-5-carboxypentyl) phthalate (5cx-MEPP), mono (2-(carboxylmethyl)hexyl) phthalate (2cx-MMHP), mono-n-octyl-phthalate (MnOP), mono-iso-nonyl phthalate (MiNP), mono-(3-carboxypropyl) phthalate (MCPP), mono-iso-decyl phthalate (MiDP), mono-ethyl phthalate (MEP), mono-cyclohexyl phthalate (MCHP), mono-methyl phthalate (MMP), mono-n-pentyl phthalate (MnPP), and their respective 13C4 or 13C2-labeled internal standards were purchased from Cambridge Isotope Laboratory (Cambridge Isotope Laboratory, Cambridge, MA, USA).
The limits of detection (LOD) ranged from 0.01 ng/mL (MCHP) to 2.3 ng/mL (2cx-MMHP). For each instrumental run, reagent blanks and QC samples were included for quality control. The method accuracies were between 80% and 120%, and precisions were lower than 10% relative standard deviation (RSD) for high and low concentrations of spiked samples (n = 7).

2.4. Statistical Analysis

We summarized the distribution of urinary phthalate metabolites using median and interquartile range. For statistical analysis, Chi-square, Fisher’s exact, and Independent Samples Test were performed using SPSS statistical software (version 14.0, SPSS Inc., Chicago, IL, USA). Phthalate metabolites that were detected at less than 75% were excluded from further analyses. All of the phthalate metabolite data were assessed by Kolmogorov-Smirnov test to evaluate whether they are normally distributed. Since the distribution of phthalate metabolite concentrations was most likely right skewed, all the phthalate metabolite measurements were logarithm transformed to approximate normal distribution. The total concentration of phthalate metabolites was creatinine standardized using the following formula to address urinary dilution as stated in previous studies: Total (phthalate metabolites) = 100 × phthalate (ng/mL)/creatinine (mg/dL) [29,30].
DEHP is rapidly metabolized to its monoester, MEHP, which is further extensively modified by various secondary metabolites, including MEHHP, MEOHP, 5cx-MEPP, and 2cx-MMHP. Therefore, to determine total exposure to DEHP, we calculated the urinary level of these metabolites by summing the molar concentrations of MEHP, MEHHP, and MEOHP (∑3-DEHP) as previously described (∑3-DEHP = (MEHP × (1/278.34)) + (MEHHP × (1/294.34)) + (MEOHP × (1/292.33)) nmol/mL) [31,32]. We also calculated the molar sum of 4-DEHP and 5-DEHP by combining the levels of ∑3-DEHP plus 5cx-MEPP (∑4-DEHP = ∑3-DEHP + (5cx-MEPP × (1/308.33)) nmol/mL), and 2cx-MMHP (∑5-DEHP = ∑4-DEHP + (2cx-MMHP × (1/308.33)) nmol/mL), respectively [33,34,35]. In addition, since 5cx-MEPP and 2cx-MMHP are oxidation products with relatively longer elimination half-lives (15–24 h) than those of other DEHP metabolites (approximately 5–10 h) [36], we further calculated the molar sum of 5cx-MEPP and 2cx-MMHP (∑2-DEHP) to determine relatively longer term human exposures to DEHP.
Multiple logistic regression analyses were performed to estimate odds ratios to determine the association between leiomyoma and the concentration of each urinary phthalate metabolite or the molar sum of DEHP metabolites. The concentration of urinary phthalate metabolites and the molar sum of DEHP metabolites were analyzed with a separate logistic model due to a possible correlation with each other. Covariates for adjustment during the multiple logistic regression were determined based on their significance after univariate analyses (p < 0.15); however, BMI was excluded since it is highly correlated with waist circumference. p values less than 0.05 were considered statistically significant.

3. Results

The demographic characteristics of the controls and cases with uterine leiomyoma are shown in Table 1. There were no differences in dysmenorrhea, smoking, exposure to second hand smoke, and alcohol consumption between the two groups; however, age, waist circumference, and parity were higher in the leiomyoma group than the control group.
Sixteen phthalate metabolites were detected in similar proportion in both the control and leiomyoma group. MnOP, MnPP, MiDP, and MiNP were rarely detected. MCHP was detected at a rate of 18.5% in the control group and 16.7% in the leiomyoma group (Table 2). Therefore, we excluded seven phthalate metabolites (MnBP, MCHP, MCPP, MnOP, MnPP, MiDP, and MiNP) for further analysis because their detection levels were less than 75%.
The log transformed creatinine-adjusted levels of MEHP (1.08 ± 0.14 µg/g creatinine), MEHHP (1.52 ± 0.12 µg/g creatinine), 2cx-MMHP (1.42 ± 0.12 µg/g creatinine), ∑3-DEHP (−0.63 ± 0.12 µg/g creatinine), ∑4-DEHP (−0.46 ± 0.12 µg/g creatinine), and ∑5-DEHP (−0.35 ± 0.12 µg/g creatinine) in the leiomyoma group were significantly higher than those of controls as shown in Table 3 (p < 0.05).
When we adjusted for age, waist circumference, and parity using multiple logistic regression analyses, we found log∑3-DEHP (OR = 10.82; 95% CI = 1.25, 93.46) and log∑4-DEHP (OR = 8.78; 95% CI = 1.03, 75.29) were significantly associated with uterine leiomyoma. We also found that logMEHP, logMEHHP and log∑5-DEHP increased the odds of leiomyoma; however, the results were borderline significant (p = 0.068, 0.076, and 0.07, respectively) (Table 4). When we further analyzed multiple logistic regression after adjustment for age, waist circumference, parity, and smoking status, log∑3-DEHP (OR = 10.78; 95% CI = 1.24, 93.81) and log∑4-DEHP (OR = 8.61; 95% CI = 1.00, 74.02) were still significant correlated with uterine leiomyoma as shown in Table 4.

4. Discussion

In this study, we measured urinary levels of 16 phthalate metabolites in patients with and without uterine leiomyoma and showed that logMEHP, logMEHHP, log2cx-MMHP, log∑3-DEHP, log∑4-DEHP, and log∑5-DEHP in the leiomyoma group were significantly higher than those in the controls. Log∑3-DEHP and log∑4-DEHP levels were significant factors associated with uterine leiomyoma after multiple logistic regression analysis. LogMEHP, logMEHHP, and log∑5-DEHP also increased the odds of leiomyoma after adjusting for confounding variables; however, the results were borderline significant.
DEHP is a phthalate that is rapidly metabolized to its monoester, MEHP, and then further modified by various side-chain hydroxylation and oxidation reactions [37,38]. Primary and secondary phthalate metabolites are known to be excreted in urine as glucuronic acid conjugates. In particular, Koch and colleagues reported that 67% of the DEHP dose was excreted in urine after 24 hours and was comprised of MEHHP (23.3%), 5cx-MEPP (18.5%), MEOHP (15.0%), MEHP (5.9%) and 2cx-MMHP (4.2%) [36]. Therefore, urinary phthalate metabolites are suggested to be sensitive biomarkers for evaluating human exposures [39,40,41]. Most toxicity studies have focused on the simple monoester MEHP; however, secondary and oxidized DEHP metabolites can occur at levels 100-fold higher and are suggested to be the ultimate developmental toxicants [42]. Based on metabolism time and half-lives of MEHP, MEHHP, and MEOHP, their presence in the urine reflects short-term exposures, whereas 5cx-MEHP and 2cx-MMHP represent relatively long-term exposures [39]. The relatively long half-lives for elimination make 5cx-MEHP and 2cx-MMHP excellent parameters for measuring the time-weighted body burden of DEHP. In studies examining estrogen-dependent diseases, MEHHP and MEOHP are primarily analyzed [22]; therefore, analysis of 5cx-MEHP and 2cx-MMHP, as well as the molar sum of these metabolites (e.g., ∑4-DEHP, ∑5-DEHP, and ∑2-DEHP) is a relatively new strategy [32,33,34,35].
Since phthalates are rapidly metabolized and excreted, phthalate metabolite concentrations detected in a single urine sample represent only recent exposures to the patients. However, many studies have relied upon a single spot urine sample that may not reflect the timing of disease onset and progression. The nearly ubiquitous presence and high volume production of short-lived chemicals, such as the phthalates, suggests continual human exposure despite their short-lived nature [41,43]. Creatinine-corrected urinary concentrations of MEP have been moderately reproducible (intraclass correlation coefficient (ICC) > 0.48) across 2–4 week sampling intervals among studies in women of reproductive age [44,45]. In addition, creatinine-corrected urinary concentrations of MBzP have high temporal reliability (ICC > 0.53) across studies of various populations evaluating time intervals ranging from 8 days to 6 months [44,45,46,47,48]. Despite a lack of temporal reliability for urinary DEHP metabolites (ICC = 0.13–0.22) compared to other metabolites with longer half-lives, the data reported here suggests that a single sample may be representative of exposure levels over time, particularly if exposure is consistent [41].
Five previous studies evaluated phthalate metabolites and uterine leiomyoma. In a cross-sectional study utilizing NHANES data, Weuve et al. reported that MEHP is inversely associated with uterine leiomyoma, while MnBP levels are positively associated with self-reported uterine leiomyoma [20]. Wise et al. relied on proxy exposure assessment and self-reported outcomes [23], and Luisi et al. evaluated phthalate levels in the blood [25]; however, methodological issues were identified in both studies. Phthalates are quickly hydrolyzed to their monoesters leading to higher phthalate monoesters; therefore, phthalates in blood may not accurately reflect internal dose. As such, concentration of phthalates in urine is the best matrix to assess the level of phthalate exposure [49]. Although a small case-control study consisting of 36 leiomyoma cases and 29 controls, Huang et al. found that urinary ∑DEHP is associated with an increased risk of leiomyoma after adjusting for the GSTM1 genotype, which is the first study to explore the gene-environmental interaction of phthalate exposure and GSTM1 polymorphisms [24]. Several studies using urine samples failed to show a constant association between urinary phthalate concentrations and uterine leiomyoma [25,26]. Recently, in an operative cohort study joining 14 clinical centers and consisting of 99 cases and 374 controls, Pollack et al. did not find an association between phthalates and uterine leiomyomas [26]. However, our results show a positive association between urinary ∑DEHP and uterine leiomyoma despite a smaller sample size. These discrepancies may be explained by differences in patient recruitment methodologies; Pollack et al. recruited patients diagnosed during laparoscopy or laparotomy only. Direct surgical visualization can miss intramural, submucosal, and subserosal fibroids, the most common fibroid types. In contrast, we recruited uterine leiomyoma patients without any other medical issues, including endometriosis and/or adenomyosis, after confirming the diagnosis with gynecologic ultrasonography as well as direct visualization during operation and subsequent pathological analysis. Cases with uterine leiomyoma only had significantly higher levels of urinary DEHP metabolites (log∑3-DEHP and log∑4-DEHP) than controls, suggesting that exposure to phthalates might affect the pathogenesis of leiomyoma.
Numerous animal studies have reported that phthalates induce dysfunction of the reproductive system, mainly in male animals, by inhibiting androgen production and steroidogenesis pathways [50,51,52]. However, several in vitro and in vivo studies showed estrogenic effect of phthalates [14,53,54], although the estrogenic activities of phthalates are still controversial and may differ by species, sex, time, and dose [55,56,57,58]. Previous studies have reported estrogenic effects of DEHP in fish [53,59] and mammals [14,60,61], and these estrogenic effects are suggested to be attributable to stimulation of aromatase (CYP19) and an increase in Vtg expression, which increases estrogen levels. Furthermore, Borch et al. reported an increase in anogenital distance (AGD) in female rats [55] after in utero DiBP exposure. Another study by Borch et al. showed decreased PPARγ protein levels in prenatally DEHP-exposed female rats [62]. A study by Boberg et al. also showed similar results; DiBP increased AGD and ovarian aromatase mRNA levels and decreased hepatic gene expression of PPARα in female rats [56]. PPARs as a super family are known to be involved in various physiological pathways including reproductive hormone synthesis, fatty acid metabolism, insulin function, immunology, central nervous system functions, and cancer development [63,64]. In particular, expression of PPARγ in granulosa cells is involved in various ovarian functions [65], and activation of the aromatase enzyme, which is responsible for the final conversion of testosterone to 17β-estradiol [66], is mediated by PPARγ [67,68]. Therefore, phthalate exposure may interfere with PPARγ expression, resulting in elevated aromatase and estradiol levels.
Moreover, aromatase is stimulated by follicle-stimulating hormone (FSH) in rat, ruminants, and humans [69,70], acting mainly through the cAMP/protein kinase A [71]. An animal study by Gonzalez-Robayna et al. showed that FSH also activates protein kinase B through phosphatidylinositol 3-kinase (PI3K) in rat granulosa cells [72], and granulosa cell differentiation and aromatase expression require activation of this pathway [73,74]. Therefore, intracellular signaling by gonadotrophic hormones for stimulation of aromatase expression/activity and estradiol accumulation is a complex process involving many potential pathway interactions. Recently, a murine study by Hannon et al. showed that DEHP disrupts estrous cyclicity and accelerates primordial follicle recruitment through dysregulation of PI3K signaling [54]. DEHP was shown to selectively increase mRNA levels of the stimulatory PI3K signaling factors, including Mtorc1 and Pdpk1 [54]. Therefore, according to their findings, DEHP promotes the PI3K signaling pathway to accelerate aromatase activity for more estrogenic activities. In particular, the PI3K signaling pathway is involved in cell proliferation, survival, migration, metabolism, and ovarian folliculogenesis [75,76,77]. Furthermore, Sharma and Singh reported that PPARγ is involved in FSH and PI3K signaling pathways for developing polycystic ovarian syndrome in response to rosiglitazone, an insulin sensitizer [78]. Taken together, metabolites of DEHP may be associated with estrogen-dependent diseases such as uterine leiomyoma that result from an increase in aromatase and estradiol levels attributable to dysregulation of PI3K signaling and PPARγ activity.
In our study, we found that age and waist circumference are potential risk factors for uterine leiomyoma. The age finding is consistent with other studies [21]; however, in this study, alcohol, and dysmenorrhea were not determined to be risk factors for developing uterine leiomyoma. This may be attributed to the small sample size, which limits our interpretation and justifies conducting larger studies. LogMEHP, logMEHHP and log∑5-DEHP increased the odds of leiomyoma after multiple logistic regression analysis with borderline significance, which is likely attributable to the small sample size. Furthermore, levels of urinary DEHP metabolites (log∑3-DEHP and log∑4-DEHP) showed a significantly higher odds ratio of uterine leiomyoma after adjustment for other risk factors, including age, waist circumference, parity, and smoking; however, the small sample sized used in this study resulted in very wide confidence intervals. Consequently, the small sample size should be considered when interpreting these results and merits caution. In addition to small sample size, there are other limitations of this study. For example, we analyzed phthalate metabolites in single, spot urine samples collected after the onset of symptoms in cases with leiomyoma. The natural history of uterine leiomyoma is complex and can have notable growth in the six months prior to detection [79]. Therefore, phthalate levels measured at the time of diagnosis may reflect a relevant period at least for their short-term growth. The comparison of phthalates between cases and controls who all have medical reasons warranting surgery that could be associated with other phthalate exposures is another limitation of this study. For example, parenteral exposure to phthalates may occur through medical devices such as PVC tubes and blood bags [9]. However, we collected urine samples under the same conditions in both case and control cases prior to each operation. Therefore, the effect of parenteral exposure to phthalates would be negligible. In addition, the frequency of phthalate exposure in controls would vary depending on the selection of controls and the indications for surgery. We divided case and control groups according to the existence of myoma, and we further excluded cases with pathological evidence of adenomyosis or endometriosis. We also excluded women with a history of occupational exposure to reproductive toxicants, hormone therapy, and any malignancy or other reproductive treatment in both case and control groups to minimize this effect. Despite these precautions, our controls were diagnosed with gynecologic conditions such as benign ovarian cyst and carcinoma in situ of the uterine cervix. Although we found no evidence in the literature that phthalate exposure is associated with these diseases [80,81], we cannot exclude the possibility that the medical necessity for surgery in control cases was associated with phthalate exposure. In addition, we did not have age-matched controls for the cases with leiomyoma. Leiomyomas are generally detected upon the onset of symptoms among women in the 4th and 5th decade of life. Therefore, we used age as a covariance in the regression model. Finally, we used a hospital-based control group instead of a population-based control group. However, other studies have also relied on a patient control group without uterine leiomyoma.

5. Conclusions

Despite these limitations, we found a significant increase in the risk of uterine leiomyoma in subjects with higher levels of total urinary DEHP metabolites (log∑3-DEHP and log∑4-DEHP) and increased age. Our findings suggest that there is an association between phthalate exposure and uterine leiomyoma. However, larger studies are warranted.

Acknowledgments

The authors thank all the participants of the study and our study team for their contributions. This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

Author Contributions

Young Ah Kim, Kyoung Chul Chun, Jae Whoan Koh, Jeong Woo Park and Yoon Hee Cho conceived, designed, and collected samples from patients; Younglim Kho performed the experiment to measure phthalate levels; Young Ah Kim, Melisa Bunderson-Schelvan and Yoon Hee Cho analyzed the data, wrote and revised the paper.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Wilcox, L.S.; Koonin, L.M.; Pokras, R.; Strauss, L.T.; Xia, Z.; Peterson, H.B. Hysterectomy in the United States, 1988–1990. Obstet. Gynecol. 1994, 83, 549–555. [Google Scholar] [CrossRef] [PubMed]
  2. Farquhar, C.M.; Steiner, C.A. Hysterectomy rates in the United States 1990–1997. Obstet. Gynecol. 2002, 99, 229–234. [Google Scholar] [CrossRef] [PubMed]
  3. Devlieger, R.; D’Hooghe, T.; Timmerman, D. Uterine adenomyosis in the infertility clinic. Hum. Reprod. Update 2003, 9, 139–147. [Google Scholar] [CrossRef] [PubMed]
  4. Guo, S.W. Glutathione S-transferases M1/T1 gene polymorphisms and endometriosis: A meta-analysis of genetic association studies. Mol. Hum. Reprod. 2005, 11, 729–743. [Google Scholar] [CrossRef] [PubMed]
  5. Baird, D.D.; Dunson, D.B.; Hill, M.C.; Cousins, D.; Schectman, J.M. High cumulative incidence of uterine leiomyoma in black and white women: Ultrasound evidence. Am. J. Obstet. Gynecol. 2003, 188, 100–107. [Google Scholar] [CrossRef] [PubMed]
  6. Gray, T.J.; Gangolli, S.D. Aspects of the testicular toxicity of phthalate esters. Environ. Health Perspect. 1986, 65, 229–235. [Google Scholar] [CrossRef] [PubMed]
  7. Kluwe, W.M. Overview of phthalate ester pharmacokinetics in mammalian species. Environ. Health Perspect. 1982, 45, 3–9. [Google Scholar] [CrossRef] [PubMed]
  8. David, R.M. Exposure to phthalate esters. Environ. Health Perspect. 2000, 108, A440. [Google Scholar] [CrossRef] [PubMed]
  9. Hashizume, K.; Nanya, J.; Toda, C.; Yasui, T.; Nagano, H.; Kojima, N. Phthalate esters detected in various water samples and biodegradation of the phthalates by microbes isolated from river water. Biol. Pharm. Bull. 2002, 25, 209–214. [Google Scholar] [CrossRef] [PubMed]
  10. Wittassek, M.; Wiesmuller, G.A.; Koch, H.M.; Eckard, R.; Dobler, L.; Muller, J.; Angerer, J.; Schluter, C. Internal phthalate exposure over the last two decades—A retrospective human biomonitoring study. Int. J. Hyg. Environ. Health 2007, 210, 319–333. [Google Scholar] [CrossRef] [PubMed]
  11. Harris, C.A.; Henttu, P.; Parker, M.G.; Sumpter, J.P. The estrogenic activity of phthalate esters in vitro. Environ. Health Perspect. 1997, 105, 802–811. [Google Scholar] [CrossRef] [PubMed]
  12. Jin, Q.; Sun, Z.; Li, Y. Estrogenic activities of di-2-ethylhexyl phthalate. Front. Med. China 2008, 2, 303–308. [Google Scholar] [CrossRef]
  13. Okubo, T.; Suzuki, T.; Yokoyama, Y.; Kano, K.; Kano, I. Estimation of estrogenic and anti-estrogenic activities of some phthalate diesters and monoesters by MCF-7 cell proliferation assay in vitro. Biol. Pharm. Bull. 2003, 26, 1219–1224. [Google Scholar] [CrossRef] [PubMed]
  14. Ma, M.; Kondo, T.; Ban, S.; Umemura, T.; Kurahashi, N.; Takeda, M.; Kishi, R. Exposure of prepubertal female rats to inhaled di(2-ethylhexyl)phthalate affects the onset of puberty and postpubertal reproductive functions. Toxicol. Sci. Off. J. Soc. Toxicol. 2006, 93, 164–171. [Google Scholar] [CrossRef] [PubMed]
  15. Moore, N.P. The oestrogenic potential of the phthalate esters. Reprod. Toxicol. 2000, 14, 183–192. [Google Scholar] [CrossRef]
  16. Roy, D.; Morgan, M.; Yoo, C.; Deoraj, A.; Roy, S.; Yadav, V.K.; Graoub, M.; Assaggaf, H.; Doke, M. Integrated bioinformatics, environmental epidemiologic and genomic approaches to identify environmental and molecular links between endometriosis and breast cancer. Int. J. Mol. Sci. 2015, 16, 25285–25322. [Google Scholar] [CrossRef] [PubMed]
  17. Kim, S.H.; Cho, S.; Ihm, H.J.; Oh, Y.S.; Heo, S.H.; Chun, S.; Im, H.; Chae, H.D.; Kim, C.H.; Kang, B.M. Possible role of phthalate in the pathogenesis of endometriosis: In vitro, animal, and human data. J. Clin. Endocrinol. Metab. 2015, 100, E1501–E1511. [Google Scholar] [CrossRef] [PubMed]
  18. Upson, K.; Sathyanarayana, S.; De Roos, A.J.; Thompson, M.L.; Scholes, D.; Dills, R.; Holt, V.L. Phthalates and risk of endometriosis. Environ. Res. 2013, 126, 91–97. [Google Scholar] [CrossRef] [PubMed]
  19. Baird, D.D.; Newbold, R. Prenatal diethylstilbestrol (DES) exposure is associated with uterine leiomyoma development. Reprod. Toxicol. 2005, 20, 81–84. [Google Scholar] [CrossRef] [PubMed]
  20. Weuve, J.; Hauser, R.; Calafat, A.M.; Missmer, S.A.; Wise, L.A. Association of exposure to phthalates with endometriosis and uterine leiomyomata: findings from NHANES, 1999–2004. Environ. Health Perspect. 2010, 118, 825–832. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Hodges, L.C.; Bergerson, J.S.; Hunter, D.S.; Walker, C.L. Estrogenic effects of organochlorine pesticides on uterine leiomyoma cells in vitro. Toxicol. Sci. Off. J. Soc. Toxicol. 2000, 54, 355–364. [Google Scholar] [CrossRef]
  22. Huang, P.C.; Li, W.F.; Liao, P.C.; Sun, C.W.; Tsai, E.M.; Wang, S.L. Risk for estrogen-dependent diseases in relation to phthalate exposure and polymorphisms of CYP17A1 and estrogen receptor genes. Environ. Sci. Pollut. Res. Int. 2014, 21, 13964–13973. [Google Scholar] [CrossRef] [PubMed]
  23. Wise, L.A.; Palmer, J.R.; Reich, D.; Cozier, Y.C.; Rosenberg, L. Hair relaxer use and risk of uterine leiomyomata in African-American women. Am. J. Epidemiol. 2012, 175, 432–440. [Google Scholar] [CrossRef] [PubMed]
  24. Huang, P.C.; Tsai, E.M.; Li, W.F.; Liao, P.C.; Chung, M.C.; Wang, Y.H.; Wang, S.L. Association between phthalate exposure and glutathione S-transferase M1 polymorphism in adenomyosis, leiomyoma and endometriosis. Hum. Reprod. 2010, 25, 986–994. [Google Scholar] [CrossRef] [PubMed]
  25. Luisi, S.; Latini, G.; de Felice, C.; Sanseverino, F.; di Pasquale, D.; Mazzeo, P.; Petraglia, F. Low serum concentrations of di-(2-ethylhexyl)phthalate in women with uterine fibromatosis. Gynecol. Endocrinol. Off. J. Int. Soc. Gynecol. Endocrinol. 2006, 22, 92–95. [Google Scholar] [CrossRef] [PubMed]
  26. Pollack, A.Z.; Buck Louis, G.M.; Chen, Z.; Sun, L.; Trabert, B.; Guo, Y.; Kannan, K. Bisphenol A, benzophenone-type ultraviolet filters, and phthalates in relation to uterine leiomyoma. Environ. Res. 2015, 137, 101–107. [Google Scholar] [CrossRef] [PubMed]
  27. Myers, S.L.; Baird, D.D.; Olshan, A.F.; Herring, A.H.; Schroeder, J.C.; Nylander-French, L.A.; Hartmann, K.E. Self-report versus ultrasound measurement of uterine fibroid status. J. Women’s Health 2012, 21, 285–293. [Google Scholar] [CrossRef] [PubMed]
  28. Kho, Y.L.; Jeoung, J.Y.; Choi, K.H.; Kim, P.G. Determination of phthalate metabolites in Korean children’s urine by high performance liquid chromatography with triple quadrupole tandem mass spectrometry. J. Environ. Health Sci. 2008, 34, 271–278. [Google Scholar] [CrossRef]
  29. Cline, R.E.; Hill, R.H., Jr.; Phillips, D.L.; Needham, L.L. Pentachlorophenol measurements in body fluids of people in log homes and workplaces. Arch. Environ. Contam. Toxicol. 1989, 18, 475–481. [Google Scholar] [CrossRef] [PubMed]
  30. Shealy, D.B.; Barr, J.R.; Ashley, D.L.; Patterson, D.G., Jr.; Camann, D.E.; Bond, A.E. Correlation of environmental carbaryl measurements with serum and urinary 1-naphthol measurements in a farmer applicator and his family. Environ. Health Perspect. 1997, 105, 510–513. [Google Scholar] [CrossRef] [PubMed]
  31. Zhao, Y.; Shi, H.J.; Xie, C.M.; Chen, J.; Laue, H.; Zhang, Y.H. Prenatal phthalate exposure, infant growth, and global DNA methylation of human placenta. Environ. Mol. Mutagen. 2015, 56, 286–292. [Google Scholar] [CrossRef] [PubMed]
  32. Barrett, E.S.; Parlett, L.E.; Sathyanarayana, S.; Redmon, J.B.; Nguyen, R.H.; Swan, S.H. Prenatal stress as a modifier of associations between phthalate exposure and reproductive development: Results from a Multicentre Pregnancy Cohort Study. Paediatr. Perinat. Epidemiol. 2016, 30, 105–114. [Google Scholar] [CrossRef] [PubMed]
  33. Messerlian, C.; Souter, I.; Gaskins, A.J.; Williams, P.L.; Ford, J.B.; Chiu, Y.; Calafat, A.M.; Hauser, R.; Earth Study Team. Urinary phthalate metabolites and ovarian reseve among women seeking infertility care. Hum. Reprod. 2016, 31, 75–83. [Google Scholar] [CrossRef] [PubMed]
  34. Alur, S.; Wang, H.; Hoeger, K.; Swan, S.H.; Sathyanarayana, S.; Redmon, B.; Nguyen, R.; Barrett, E.S. Urinary phthalate metabolites concentrations in relation to history of infertility and use of assisted reproductive technology. Fertil. Steril. 2016, 104, 1227–1235. [Google Scholar] [CrossRef] [PubMed]
  35. Herr, C.; zur Nieden, A.; Koch, H.M.; Schuppe, H.C.; Fieber, C.; Angerer, J.; Eikmann, T.; Stillanakis, N.I. Urinary di(2-ethylhexyl) phthalate (DEHP)—Metabolites and male human markers of reproductive function. Int. J. Hyg. Environ. Health 2009, 212, 648–653. [Google Scholar] [CrossRef] [PubMed]
  36. Koch, H.M.; Bolt, H.M.; Preuss, R.; Angerer, J. New metabolites of di(2-ethylhexyl)phthalate (DEHP) in human urine and serum after single oral doses of deuterium-labelled DEHP. Arch. Toxicol. 2005, 79, 367–376. [Google Scholar] [CrossRef] [PubMed]
  37. Albro, P.W.; Corbett, J.T.; Schroeder, J.L.; Jordan, S.; Matthews, H.B. Pharmacokinetics, interactions with macromolecules and species differences in metabolism of DEHP. Environ. Health Perspect. 1982, 45, 19–25. [Google Scholar] [CrossRef] [PubMed]
  38. Schmid, P.; Schlatter, C. Excretion and metabolism of di(2-ethylhexyl)-phthalate in man. Xenobiotica 1985, 15, 251–256. [Google Scholar] [CrossRef] [PubMed]
  39. Becker, K.; Goen, T.; Seiwert, M.; Conrad, A.; Pick-Fuss, H.; Muller, J.; Wittassek, M.; Schulz, C.; Kolossa-Gehring, M. GerES IV: Phthalate metabolites and bisphenol A in urine of German children. Int. J. Hyg. Environ. Health 2009, 212, 685–692. [Google Scholar] [CrossRef] [PubMed]
  40. Frederiksen, H.; Aksglaede, L.; Sorensen, K.; Skakkebaek, N.E.; Juul, A.; Andersson, A.M. Urinary excretion of phthalate metabolites in 129 healthy Danish children and adolescents: Estimation of daily phthalate intake. Environ. Res. 2011, 111, 656–663. [Google Scholar] [CrossRef] [PubMed]
  41. Silva, M.J.; Barr, D.B.; Reidy, J.A.; Malek, N.A.; Hodge, C.C.; Caudill, S.P.; Brock, J.W.; Needham, L.L.; Calafat, A.M. Urinary levels of seven phthalate metabolites in the U.S. population from the National Health and Nutrition Examination Survey (NHANES) 1999–2000. Environ. Health Perspect. 2004, 112, 331–338. [Google Scholar] [CrossRef] [PubMed]
  42. Regnier, J.; Bowden, C.; Lhuguenot, J. Effects on rat embryonic development in vitro of di-(2-ethylhexyl) phthalate (DEHP) and its metabolites. Toxicologist 2004, 78, 38–39. [Google Scholar]
  43. Barr, D.B.; Silva, M.J.; Kato, K.; Reidy, J.A.; Malek, N.A.; Hurtz, D.; Sadowski, M.; Needham, L.L.; Calafat, A.M. Assessing human exposure to phthalates using monoesters and their oxidized metabolites as biomarkers. Environ. Health Perspect. 2003, 111, 1148–1151. [Google Scholar] [CrossRef] [PubMed]
  44. Baird, D.D.; Saldana, T.M.; Nepomnaschy, P.A.; Hoppin, J.A.; Longnecker, M.P.; Weinberg, C.R.; Wilcox, A.J. Within-person variability in urinary phthalate metabolite concentrations: Measurements from specimens after long-term frozen storage. J. Expo. Sci. Environ. Epidemiol. 2010, 20, 169–175. [Google Scholar] [CrossRef] [PubMed]
  45. Peck, J.D.; Sweeney, A.M.; Symanski, E.; Gardiner, J.; Silva, M.J.; Calafat, A.M.; Schantz, S.L. Intra-and inter-individual variability of urinary phthalate metabolite concentrations in Hmong women of reproductive age. J. Expo. Sci. Environ. Epidemiol. 2010, 20, 90–100. [Google Scholar] [CrossRef] [PubMed]
  46. Adibi, J.J.; Whyatt, R.M.; Williams, P.L.; Calafat, A.M.; Camann, D.; Herrick, R.; Nelson, H.; Bhat, H.K.; Perera, F.P.; Silva, M.J. Characterization of phthalate exposure among pregnant women assessed by repeat air and urine samples. Environ. Health Perspect. 2008, 116, 467–473. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Fromme, H.; Bolte, G.; Koch, H.M.; Angerer, J.; Boehmer, S.; Drexler, H.; Mayer, R.; Liebl, B. Occurrence and daily variation of phthalate metabolites in the urine of an adult population. Int. J. Hyg. Environ. Health 2007, 210, 21–33. [Google Scholar] [CrossRef] [PubMed]
  48. Teitelbaum, S.; Britton, J.; Calafat, A.; Ye, X.; Silva, M.; Reidy, J.; Galvez, M.; Brenner, B.; Wolff, M. Temporal variability in urinary concentrations of phthalate metabolites, phytoestrogens and phenols among minority children in the United States. Environ. Res. 2008, 106, 257–269. [Google Scholar] [CrossRef] [PubMed]
  49. Calafat, A.M.; Koch, H.M.; Swan, S.H.; Hauser, R.; Goldman, L.R.; Lanphear, B.P.; Longnecker, M.P.; Rudel, R.A.; Teitelbaum, S.L.; Whyatt, R.M.; et al. Misuse of blood serum to assess exposure to bisphenol A and phthalates. Breast Cancer Res. 2013, 15, 403. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Barlow, N.J.; Phillips, S.L.; Wallace, D.G.; Sar, M.; Gaido, K.W.; Foster, P.M. Quantitative changes in gene expression in fetal rat testes following exposure to di(n-butyl)phthalate. Toxicol. Sci. 2003, 73, 431–441. [Google Scholar] [CrossRef] [PubMed]
  51. Lehmann, K.P.; Phillips, S.; Sar, M.; Foster, P.M.; Gaido, K.W. Dose-dependent alterations in gene expression and testosterone synthesis in the fetal testes of male rats exposed to di(n-butyl)phthalate. Toxicol. Sci. 2004, 81, 60–68. [Google Scholar] [CrossRef] [PubMed]
  52. Howdeshell, K.L.; Wilson, V.S.; Furr, J.; Lambright, C.R.; Rider, C.V.; Blystone, C.R.; Hotchkiss, A.K.; Gray, L.E. A mixture of five phthalate esters inhibits fetal testicular testosterone production in the sprague-dawley rat in a cumulative, dose-additive manner. Toxicol. Sci. 2008, 105, 153–165. [Google Scholar] [CrossRef] [PubMed]
  53. Wang, X.; Yang, Y.; Zhang, L.; Ma, Y.; Han, J.; Yang, L.; Zhou, B. Endocrine disruption by di-(2-ethylhexyl)-phthalate in Chinese rare minnow (Gobiocypris rarus). Environ. Toxicol. Chem. 2013, 32, 1846–1854. [Google Scholar] [CrossRef] [PubMed]
  54. Hannon, P.R.; Peretz, J.; Flaws, J.A. Daily exposure to di(2-ethylhexyl) phthalate alters estrous cyclicity and accelerates primordial follicle recruitment potentially via dysregulation of the phosphatidylinositol 3-kinase signaling pathway in adult mice. Biol. Reprod. 2014, 90, 136. [Google Scholar] [CrossRef] [PubMed]
  55. Borch, J.; Axelstad, M.; Vinggaard, A.M.; Dalgaard, M. Diisobutyl phthalate has comparable anti-androgenic effects to di-n-butyl phthalate in fetal rat testis. Toxicol. Lett. 2006, 163, 183–190. [Google Scholar] [CrossRef] [PubMed]
  56. Boberg, J.; Metzdorff, S.; Wortziger, R.; Axelstad, M.; Brokken, L.; Vinggaard, A.M.; Dalgaard, M.; Nellemann, C. Impact of diisobutyl phthalate and other PPAR agonists on steroidogenesis and plasma insulin and leptin levels in fetal rats. Toxicology 2008, 250, 75–81. [Google Scholar] [CrossRef] [PubMed]
  57. Grande, S.W.; Andrade, A.J.; Talsness, C.E.; Grote, K.; Golombiewski, A.; Sterner-Kock, A.; Chahoud, I. A dose-response study following in utero and lactational exposure to di-(2-ethylhexyl) phthalate (DEHP): Reproductive effects on adult female offspring rats. Toxicology 2007, 229, 114–122. [Google Scholar] [CrossRef] [PubMed]
  58. Rider, C.V.; Furr, J.; Wilson, V.S.; Gray, L.E. A mixture of seven antiandrogens induces reproductive malformations in rats. Int. J. Androl. 2008, 31, 249–262. [Google Scholar] [CrossRef] [PubMed]
  59. Ye, T.; Kang, M.; Huang, Q.; Fang, C.; Chen, Y.; Shen, H.; Dong, S. Exposure to DEHP and MEHP from hatching to adulthood causes reproductive dysfunction and endocrine disruption in marine medaka (Oryzias melastigma). Aquat. Toxicol. 2014, 146, 115–126. [Google Scholar] [CrossRef] [PubMed]
  60. Akingbemi, B.T.; Ge, R.; Klinefelter, G.R.; Zirkin, B.R.; Hardy, M.P. Phthalate-induced Leydig cell hyperplasia is associated with multiple endocrine disturbances. Proc. Natl. Acad. Sci. USA 2004, 101, 775–780. [Google Scholar] [CrossRef] [PubMed]
  61. Andrade, A.J.; Grande, S.W.; Talsness, C.E.; Grote, K.; Chahoud, I. A dose-response study following in utero and lactational exposure to di-(2-ethylhexyl)-phthalate (DEHP): Non-monotonic dose-response and low dose effects on rat brain aromatase activity. Toxicology 2006, 227, 185–192. [Google Scholar] [CrossRef] [PubMed]
  62. Borch, J.; Metzdorff, S.B.; Vinggaard, A.M.; Brokken, L.; Dalgaard, M. Mechanisms underlying the anti-androgenic effects of diethylhexyl phthalate in fetal rat testis. Toxicology 2006, 223, 144–155. [Google Scholar] [CrossRef] [PubMed]
  63. Lehrke, M.; Lazar, M.A. The many faces of PPARγ. Cell 2005, 123, 993–999. [Google Scholar] [CrossRef] [PubMed]
  64. Sharma, I.; Singh, D. Conjugated linoleic acids attenuate FSH-and IGF1-stimulated cell proliferation; IGF1, GATA4, and aromatase expression; and estradiol-17β production in buffalo granulosa cells involving PPARγ, PTEN, and PI3K/Akt. Reproduction 2012, 144, 373–383. [Google Scholar] [CrossRef] [PubMed]
  65. Sharma, I.; Monga, R.; Singh, N.; Datta, T.K.; Singh, D. Ovary-specific novel peroxisome proliferator activated receptors-gamma transcripts in buffalo. Gene 2012, 504, 245–252. [Google Scholar] [CrossRef] [PubMed]
  66. Hilscherova, K.; Jones, P.D.; Gracia, T.; Newsted, J.L.; Zhang, X.; Sanderson, J.; Richard, M.; Wu, R.S.; Giesy, J.P. Assessment of the effects of chemicals on the expression of ten steroidogenic genes in the H295R cell line using real-time PCR. Toxicol. Sci. 2004, 81, 78–89. [Google Scholar] [CrossRef] [PubMed]
  67. Lovekamp-Swan, T.; Jetten, A.M.; Davis, B.J. Dual activation of PPARα and PPARγ by mono-(2-ethylhexyl) phthalate in rat ovarian granulosa cells. Mol. Cell. Endocrinol. 2003, 201, 133–141. [Google Scholar] [CrossRef]
  68. Zaree, M.; Shahnazi, V.; Fayezi, S.; Darabi, M.; Mehrzad-Sadaghiani, M.; Darabi, M.; Khani, S.; Nouri, M. Expression levels of PPARγ and CYP-19 in polycystic ovarian syndrome primary granulosa cells: Influence of ω-3 fatty acid. Int. J. Fertil. Steril. 2015, 9, 197–204. [Google Scholar] [PubMed]
  69. Steinkampf, M.P.; Mendelson, C.R.; Simpson, E.R. Regulation by follicle-stimulating hormone of the synthesis of aromatase cytochrome P-450 in human granulosa cells. Mol. Endocrinol. 1987, 1, 465–471. [Google Scholar] [CrossRef] [PubMed]
  70. Fitzpatrick, S.L.; Richards, J.S. Regulation of Cytochrome P450 Aromatase Messenger Ribonucleic Acid and Activity by Steroids and Gonadotropins in Rat Granulosa Cells. Endocrinology 1991, 129, 1452–1462. [Google Scholar] [CrossRef] [PubMed]
  71. Silva, J.; Hamel, M.; Sahmi, M.; Price, C. Control of oestradiol secretion and of cytochrome P450 aromatase messenger ribonucleic acid accumulation by FSH involves different intracellular pathways in oestrogenic bovine granulosa cells in vitro. Reproduction 2006, 132, 909–917. [Google Scholar] [CrossRef] [PubMed]
  72. Gonzalez-Robayna, I.J.; Falender, A.E.; Ochsner, S.; Firestone, G.L.; Richards, J.S. Follicle-stimulating hormone (FSH) stimulates phosphorylation and activation of protein kinase B (PKB/Akt) and serum and glucocorticoid-induced kinase (Sgk): Evidence for A kinase-independent signaling by FSH in granulosa cells. Mol. Endocrinol. 2000, 14, 1283–1300. [Google Scholar] [CrossRef] [PubMed]
  73. Zeleznik, A.J.; Saxena, D.; Little-Ihrig, L. Protein kinase B is obligatory for follicle-stimulating hormone-induced granulosa cell differentiation. Endocrinology 2003, 144, 3985–3994. [Google Scholar] [CrossRef] [PubMed]
  74. Alam, H.; Maizels, E.T.; Park, Y.; Ghaey, S.; Feiger, Z.J.; Chandel, N.S.; Hunzicker-Dunn, M. Follicle-stimulating hormone activation of hypoxia-inducible factor-1 by the phosphatidylinositol 3-kinase/AKT/Ras homolog enriched in brain (Rheb)/mammalian target of rapamycin (mTOR) pathway is necessary for induction of select protein markers of follicular differentiation. J. Biol. Chem. 2004, 279, 19431–19440. [Google Scholar] [PubMed]
  75. Adhikari, D.; Liu, K. Molecular mechanisms underlying the activation of mammalian primordial follicles. Endocr. Rev. 2009, 30, 438–464. [Google Scholar] [CrossRef] [PubMed]
  76. Engelman, J.A.; Luo, J.; Cantley, L.C. The evolution of phosphatidylinositol 3-kinases as regulators of growth and metabolism. Nat. Rev. Genet. 2006, 7, 606–619. [Google Scholar] [CrossRef] [PubMed]
  77. Stokoe, D. The phosphoinositide 3-kinase pathway and cancer. Expert Rev. Mol. Med. 2005, 7, 1–22. [Google Scholar] [CrossRef] [PubMed]
  78. Sharma, I.; Singh, D. Rosiglitazone suppresses FSH and IGF-1 induced downstream PI3K/Akt pathway mediated granulosa cell function involving PPARg and PTEN: Potential targets of fertility regulation. Inflamm. Cell Signal. 2014, 1. [Google Scholar] [CrossRef]
  79. Peddada, S.D.; Laughlin, S.K.; Miner, K.; Guyon, J.-P.; Haneke, K.; Vahdat, H.L.; Semelka, R.C.; Kowalik, A.; Armao, D.; Davis, B.; et al. Growth of uterine leiomyomata among premenopausal black and white women. Proc. Natl. Acad. Sci. USA 2008, 105, 19887–19892. [Google Scholar] [CrossRef] [PubMed]
  80. Costa, E.M.F.; Spritzer, P.M.; Hohl, A.; Bachega, T.A. Effects of endocrine disruptors in the development of the female reproductive tract. Arq. Bras. Endocrinol. Metabol. 2014, 58, 153–161. [Google Scholar] [CrossRef] [PubMed]
  81. Kay, V.R.; Chambers, C.; Foster, W.G. Reproductive and developmental effects of phthalate diesters in females. Crit. Rev. Toxicol. 2013, 43, 200–219. [Google Scholar] [CrossRef] [PubMed]
Table 1. Clinical characteristics of cases with uterine leiomyoma and controls.
Table 1. Clinical characteristics of cases with uterine leiomyoma and controls.
VariableControl (n = 27)Cases (n = 30)p-Value
Age (years)34.78 ± 1.9042.67 ± 1.02<0.001
BMI (kg/m2)22.06 ± 0.6823.06 ± 0.540.08
Waist circumference (cm)70.06 ± 1.0674.44 ± 1.300.01
Gravida
 08 (29.63)5 (16.67)0.49
 1–210 (37.04)12 (40.0)
 3 and more9 (33.33)13 (43.33)
Parity
 012 (44.44)7 (23.33)0.02
 17 (25.93)3 (10)
 2 and more8 (29.63)20 (66.67)
Dysmenorrhea *9 (33.33)12 (40)0.60
Cigarette smoking *9 (33.33)5 (16.67)0.14
Second-hand smoking *10 (37.04)9 (30)0.36
Alcohol consumption *26 (96.29)29 (96.67)0.94
The data are presented as the mean ± SE or numbers (%); p values were calculated by t-test or x2-test. Fisher’s exact test was used when individual cells were <5; * indicates “Yes”.
Table 2. Laboratory measurement of urinary phthalate metabolite concentrations and distribution by case status.
Table 2. Laboratory measurement of urinary phthalate metabolite concentrations and distribution by case status.
Phthalate MetaboliteControlCases
Measured > LOQ
n (%)
Median (IQR)
(µg/g Creatinine)
Measured > LOQ
n (%)
Median (IQR)
(µg/g Creatinine)
MEHP26 (96.29)4 (2.68–7.66)28 (93.33)5 (2.54–29.25)
MEOHP27 (100)6.16 (4.48–10.09)30 (100)8.55 (6.14–18.35)
MEHHP27 (100)16.74 (11.63–25.36)30 (100)17.62 (13.76–37.81)
2cx-MMHP27 (100)12.89 (10.42–19.87)30 (100)17.59 (11.34–34.06)
5cx-MEPP27 (100)16.21 (12.74–22.84)30 (100)17.94 (12.24–34.40)
MnBP13 (48.14)0 (0–12.63)21 (70.00)11.62 (0–19.23)
MBzP25 (92.59)4.7 (2.16–7.83)26 (86.67)5.07 (1.55–10.08)
MiBP27 (100)4.11 (2.37–7.50)30 (100)3.93 (2.61–6.14)
MEP24 (88.89)4.87 (2.42–12.10)25 (83.33)3.96 (1.38–14.60)
MMP24 (88.89)4.18 (1.55–6.64)26 (86.67)3.98 (1.06–16.69)
MCHP5 (18.52)NA5 (16.67)NA
MnOP0NA1 (3.33)NA
MCPP19 (70.37)1.61 (0–4.01)16 (53.33)1.46 (0–6.31)
MnPP0NA1 (3.33)NA
MiDP0NA0NA
MiNP0NA0NA
LOQ = limit of quantitation; IQR = interquartile range; NA = Not available.
Table 3. The log transformed creatinine-adjusted urinary phthalate levels in cases and controls.
Table 3. The log transformed creatinine-adjusted urinary phthalate levels in cases and controls.
Urine Level (µg/g Creatinine)Control (n = 27)Cases (n = 30)p-Value
MEHP0.68 ± 0.061.08 ± 0.140.02
MEOHP0.91 ± 0.051.17 ± 0.130.08
MEHHP1.23 ± 0.051.52 ± 0.120.04
2cx-MMHP1.16 ± 0.041.42 ± 0.120.04
5cx-MEPP1.24 ± 0.041.47 ± 0.120.11
∑3-DEHP *−0.98 ± 0.05−0.63 ± 0.120.01
∑4-DEHP **−0.79 ± 0.04−0.46 ± 0.120.02
∑5-DEHP ***−0.67 ± 0.04−0.35 ± 0.120.02
∑2-DEHP ****−0.97 ± 0.03−0.73 ± 0.120.07
MBzP0.68 ± 0.070.69 ± 0.080.95
MiBP0.60 ± 0.070.64 ± 0.060.66
MEP0.83 ± 0.090.85 ± 0.130.87
MMP0.60 ± 0.090.74 ± 0.140.40
All phthalate metabolites were creatinine (mg/dL) standardized. The data are presented as the mean ± SE; * ∑3-DEHP is the molar sum of MEHP, MEHHP, and MEOHP; ** ∑4-DEHP is the molar sum of MEHP, MEHHP, MEOHP, and 5cx-MEPP; *** ∑5-DEHP is the molar sum of MEHP, MEHHP, MEOHP, 5cx-MEPP, and 2cx-MMHP; **** ∑2-DEHP is the molar sum of 5cx-MEPP and 2cx-MMHP.
Table 4. Odds ratio and 95% confidence interval (CI) for the association between leiomyoma and log transformed creatinine-adjusted urinary phthalate levels.
Table 4. Odds ratio and 95% confidence interval (CI) for the association between leiomyoma and log transformed creatinine-adjusted urinary phthalate levels.
Urine Level (µg/g Creatinine)Crude OR (95% CI)Adjusted OR a (95% CI)Adjusted OR b (95% CI)
MEHP3.75 (1.17–12.0)4.08 (0.89–18.60)4.17 (0.90–19.40)
MEOHP2.83 (0.81–9.87)3.82 (0.71–20.60)3.71 (0.71–20.80)
MEHHP3.63 (0.93–14.20)5.43 (0.84–35.20)5.52 (0.83–36.57)
2cx-MMHP3.87 (0.86–17.30)3.08 (0.63–15.10)3.10 (0.63–15.12)
5cx-MEPP2.76 (0.73–10.37)2.95 (0.58–15.02)2.95 (0.58–14.96)
∑3-DEHP *6.04 (1.24–29.37)10.82 (1.25–93.46)10.78 (1.24–93.81)
∑4-DEHP **5.73 (1.10–29.74)8.78 (1.03–72.29)8.61 (1.00–74.02)
∑5-DEHP ***6.18 (1.07–35.86)8.24 (0.86–79.10)8.01 (0.84–76.0)
∑2-DEHP ****3.53 (0.78–15.84)3.29 (0.58–18.63)3.30 (0.59–18.55)
MBzP1.06 (0.23–5.80)1.27 (0.16–6.81)1.17 (0.17–8.18)
MiBP1.42 (0.31–6.54)2.12 (0.29–15.42)2.15 (0.29–15.80)
MEP1.09 (0.39–3.07)1.27 (0.36–4.44)1.63 (0.39–6.75)
MMP1.51 (0.58–3.95)1.26 (0.40–3.95)1.15 (0.35–3.84)
All phthalate metabolites were creatinine (mg/dL) standardized. a Adjusted for age, waist circumference, and parity; b Adjusted for age, waist circumference, parity, and smoking status; * ∑3-DEHP is the molar sum of MEHP, MEHHP, and MEOHP; ** ∑4-DEHP is the molar sum of MEHP, MEHHP, MEOHP, and 5cx-MEPP; *** ∑5-DEHP is the molar sum of MEHP, MEHHP, MEOHP, 5cx-MEPP, and 2cx-MMHP; **** ∑2-DEHP is the molar sum of 5cx-MEPP and 2cx-MMHP.

Share and Cite

MDPI and ACS Style

Kim, Y.A.; Kho, Y.; Chun, K.C.; Koh, J.W.; Park, J.W.; Bunderson-Schelvan, M.; Cho, Y.H. Increased Urinary Phthalate Levels in Women with Uterine Leiomyoma: A Case-Control Study. Int. J. Environ. Res. Public Health 2016, 13, 1247. https://0-doi-org.brum.beds.ac.uk/10.3390/ijerph13121247

AMA Style

Kim YA, Kho Y, Chun KC, Koh JW, Park JW, Bunderson-Schelvan M, Cho YH. Increased Urinary Phthalate Levels in Women with Uterine Leiomyoma: A Case-Control Study. International Journal of Environmental Research and Public Health. 2016; 13(12):1247. https://0-doi-org.brum.beds.ac.uk/10.3390/ijerph13121247

Chicago/Turabian Style

Kim, Young Ah, Younglim Kho, Kyoung Chul Chun, Jae Whoan Koh, Jeong Woo Park, Melisa Bunderson-Schelvan, and Yoon Hee Cho. 2016. "Increased Urinary Phthalate Levels in Women with Uterine Leiomyoma: A Case-Control Study" International Journal of Environmental Research and Public Health 13, no. 12: 1247. https://0-doi-org.brum.beds.ac.uk/10.3390/ijerph13121247

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop