Abstract
Humans are ubiquitously exposed to environmental endocrine disrupting chemicals such as phthalates. Phthalates can migrate out of products and enter the human body through ingestion, inhalation, or dermal application, can have potential estrogenic/antiestrogenic and/or androgenic/antiandrogenic activity, and are involved in many diseases. As a female reproductive organ that is regulated by hormones such as estrogen, progesterone and androgen, the uterus can develop several disorders such as leiomyoma, endometriosis and abnormal bleeding. In this review, we summarize the hormone-like activities of phthalates, in vitro studies of endometrial cells exposed to phthalates, epigenetic modifications in the uterus induced by phthalate exposure, and associations between phthalate exposure and uterine disorders such as leiomyoma and endometriosis. Moreover, we also discuss the current research gaps in understanding the relationship between phthalate exposure and uterine disorders.
Introduction
Humans are commonly exposed to environmental endocrine-disrupting chemicals (EDCs), which can have hormonal-like effects on the entire body, and concerns about their e potential toxicity have been raised. Phthalates, a type of EDC, are used extensively as plasticizers or additives in medical devices, food packaging, toys, and personal care products such as cosmetics [1]. Phthalates can leak out of products, enter the human body through ingestion, inhalation, or dermal application and be rapidly metabolized and excreted into urine. Almost every person has detectable concentrations of phthalate metabolites in their urine [2], which reflects ubiquitous exposure that can impact various systems and lead to diseases [3].
Given the negative effects of phthalates on human health, researchers have established the tolerable daily intake values, defined as the daily intake amount of a chemical that has been assessed to be safe for humans on a long-term basis, and the reference dose, defined as the maximum acceptable oral dose of a toxic substance [2], 4]. A female reproductive organ regulated by the Hypothalamic-Pituitary-Ovarianaxis, the uterus is essential for embryo implantation, pregnancy maintenance, and delivery of the uterus. Various cell types (e.g., stromal cells, luminal epithelial cells) of the uterus endometrium and myometrium express the estrogen receptor (ER), progesterone receptor (PR), and androgen receptor (AR), whose levels vary during the menstrual cycle [5]. The function of the uterus is regulated primarily by hormones, including estrogen, progesterone and androgen.
Phthalates can exhibit estrogenic/antiestrogenic and/or androgenic/antiandrogenic activities, and it is important to investigate their effects on uterine function and associated disorders such as endometriosis (EMs) and fibroids. This review summarizes the following aspects: (1) hormone-like activities of phthalates, (2) in vitro studies of endometrial cells, (3) phthalate and epigenetic modifications, (4) associations between phthalates and leiomyoma, and (5) associations between phthalates and EMs. We focus on the common uterine-associated diseases and their potential links to phthalates.
Materials and methods
For this study, a scoping review of the literature was performed to identify published studies that describe hormone-like activities of phthalates, their effects on in vitro cultured endometrial cells, and the relationship between phthalate exposure and uterine diseases (e.g., leiomyoma and EMs). Full-length articles in English were collected from MEDLINE, PubMed, Google Scholar, Scopus, Web of Science, and ScienceDirect up to November 2023. We searched these databases using the following key terms: (1) “phthalate and estrogenic/antiestrogenic/androgenic/antiandrogenic activity”; (2) “phthalate and endometrial cells”, “phthalate and stromal cells”, “phthalate and endometrial mesenchymal stem/stromal cell”, and “phthalate and endometrial endothelial cells”; (3) “phthalates and uterine DNA methylation” and “phthalates and uterine and miRNA”; (4) “phthalates and uterine leiomyoma” and “phthalates and uterine fibroids”; (5) “phthalates and endometriosis” and “phthalates and adenomyosis”; (6) “phthalates and uterine histology changes”.
The studies selected met the following criteria: (1) studies that examined the effects of exposure to one or more phthalate ester or metabolites on the activity (agonistic or antagonist) of ERs and ARs; (2) experimental studies that reported in vitro, in vivo, or both in vitro and in vivo findings to identify the effects of phthalates on endometrial cells; (3) studies that described the effects of phthalates on uterine epigenetics; (4) human studies that explored the association between phthalate exposure and uterine disorders such as leiomyoma and EMs; and (5) studies that described the relationship between phthalates and uterine histology changes. Studies that met the following criteria were excluded: (1) no methods described; (2) no version available in the English language; (3) no full-text publication available; or (4) content redundancy. Phthalates and their metabolites measured in studies are listed in Table 1.
Phthalates and metabolites measured in studies.
Phthalate abbreviation | Name |
---|---|
1. DEHP | Bis-2-ethyl hexyl phthalate |
1.1 MEHP | Mono-ethyl hexyl phthalate |
1.1.1 MECPP | Mono (2-ethyl-5-carboxyphentyl) phthalate, secondary metabolite of DEHP. |
1.1.2 MCMHP | Mono-[(2-carboxymethyl) hexyl] phthalate, secondary metabolite of DEHP. |
1.1.3 MEHHP (5OHMEHP) | Mono-(2-ethyl-5-hydroxyhexyl) phthalate, secondary metabolite of DEHP. |
1.1.4 MEOHP (5oxo-MEHP) | Mono-(2-ethyl-5-oxo-hexyl) phthalate, secondary metabolite of DEHP. |
1.1.5 MECPP | Mono (2-ethyl-5-carboxypentyl) phthalate, secondary metabolite of DEHP. |
2. DOP (DnOP) | Di-n-octyl phthalate |
2.1 MOP (MnOP) | Primary metabolite of DnOP, monooctyl phthalate |
2.2 MCPP | One oxidative metabolite of DnOP, mono-(3-carboxypropyl) phthalate |
3. BBP (BBzP) | Benzyl butyl phthalate |
3.1 MBzP | Monobenzyl phthalate |
4. DCHP | Di-cyclohexyl phthalate |
4.1 MCHP | Metabolite of DCHP, monocyclohexyl phthalate |
5. DMP | |
5.1 MMP | Metabolite of DMP, 2-(Methoxycarbonyl)benzoic acid |
6. DBP, DnBP | Dibutyl phthalates (both di-n-butyl and di-isobutyl phthalates, referred to as DBP) |
6.1 MBP, MnBP | Primary metabolite of DBP, monobutyl phthalate (mono-n-butyl phthalate) |
6.2 MHBP | Metabolite of DBP, mono-hydroxybutyl phthalate |
6.3 MiBP | Primary metabolite of DBP, mono-isobutyl phthalate (MiBP) |
6.4 MHiBP | Metabolite of DBP, mono-hydroxyisobutyl phthalate |
7. DEP | Diethyl phthalate |
7.1 MEP | Primary metabolite of DEP, mono-ethyl phthalate |
8. BzBP (BBzP) | Benzylbutyl phthalate |
8.1 MBzP | Primary metabolite of BzBP, mono-benzyl phthalate |
9. DiNP | Di-isononyl phthalate |
9.1 MINP, MNP | Primary metabolite of DiNP, monoisonoyl phthalate |
9.1.1 MCNP (cxMINP) | Mono carboxyisononyl phthalate |
9.1.2 OHMINP | Mono-hydroxyisononyl phthalate |
9.1.3 MCOP | Mono carboxyisooctyl phthalate |
10. MCPPa | Mono-(3-carboxypropyl) phthalate |
11. DIBP | Diisobutyl phthalate |
11.1 MiBP | Monoisobutyl phthalate |
11.2 MHiBP | Mono-hydroxyisobutyl phthalate |
12. DHP | Dihexyl phthalate |
13. DiDP, | Diiso-decyl phthalate |
13.1 MCNP | Monocarboxynonyl phthalate |
14. DCHP | Dicyclohexyl phthalate |
15. DAP | Diallyl phthalate |
16. DPeP | Dipentyl phthalate |
17. DiHP | Diisohexyl phthalate |
18. DIHepP | Diisoheptyl phthalate |
19. DBzP | Dibenzyl phthalate |
-
aMCPP, non-specific metabolite of several phthalates; MnBP, metabolite of mono-n-butyl phthalate; DnBP, secondary metabolite of di-n-butyl phthalate; MnOP, metabolite of mono-n-octyl phthalate; DnOP, secondary metabolite of di-n-octyl phthalate.
The estrogenic/antiestrogenic and androgenic/antiandrogenic activities of phthalates
Phthalates have been shown to act as antagonists and/or agonists via one or more hormonal receptors, including the ERα, ERβ, and AR [6], 7], and these actions can disrupt the endocrine systems [8]. Several in vitro studies have used ER-binding assays, proliferation assays with MCF-7 cells, ER-dependent transcription assays, and ER-mediated reporter gene assays to investigate the estrogenic effects of certain phthalates [9], [10], [11], [12], [13], [14]. Another study used primary-cultured hepatocytes from adult male Xenopus laevis and an enzyme-linked immunosorbent assay (ELISA) to detect vitellogenin (VTG) [15]. In vivo models, such as immature rat uterotrophic and transgenic mice models with an ER-mediated luciferase (Luc) reporter gene system have been used to evaluate the estrogenic characteristic of phthalates [16], 17]. To evaluate AR agonist and antagonist activities, in vitro methods such as the AR-binding assay [18], AR-EcoScreen assay [19], and Luc reporter gene assay [9], 20], 21], and in vivo methods such as the Hershberger assay [22] have been used.
Numerous commercial substances and new chemicals entering the environment often lack data on their potential ER or AR bioactivities, and evaluation of their potential bioactivities and toxicity is urgently needed. However, applying the current validated methods of the U.S. Environmental Protection Agency (EPA) and Organization for Economic Cooperation and Development (OECD) is expensive and would take decades to complete all the testing. As an alternative, high-throughput in vitro screening (HTS) assays and computational toxicology methods have been developed to determine ER bioactivities and to identify potential AR-active chemicals rapidly and cost effectively [23], [24], [25]. Additionally, in silico methods and computational systems have been integrated to explore whether EDCs can bind to nuclear hormone receptors and to identify androgen-active chemicals [26], 27].
A recent study used a suite of in vitro assays and in silico models to identify which method is best for detecting endocrine-disrupting (ED) potential and reported that the effects on the ER and AR were similar. Researchers have suggested assessing the potential of ED effects of chemicals (including phthalates) by combining data from in vitro and in silico assessments, and t information from other sources [28].
studies have shown that phthalates can bind to the ER, PR and AR, and activate their associated pathways, through which they can exert either estrogenic/anti-estrogenic or androgenic/anti-androgenic activities. However, conflicting results between studies, and discrepancies between in vitro and in vivo studies have been observed, as shown in Table 2. For instance, whereas almost all studies have reported that butyl benzyl phthalate (BBP) exhibits estrogenic activity in in vitro assays, some studies have confirmed this action in vivo, but others have reported inconsistent results [8], [10], [11], [12], [13], [14, [29], [30], [31], [32], [33]. These discrepancies may reflect differences in the source of the test sample, solvent used (e.g., ethanol vs. DMSO), variation in the dose or cell lines used, or sensitivity of the assays.
In vitro and in vivo examination of the activities of phthalates.
Phthalates | Estrogenic | Anti-estrogenic | Androgenic | Anti-androgenic | ||
---|---|---|---|---|---|---|
In vitro | In vivo | In vitro | In vitro | In vitro | In vivo | |
DEHP | − [9], 11], 12], 30], 31], 34], + [8] | − [12], 31], + [17] | − [30], + [8], 14] | + [9] | − [35], + [9], 22] | + [35] |
MEHP | − [30] | − [30] | − [35], + [22] | |||
DCHP | + [8], 30], 31] | − [31] | − [30] | + [8] | ||
MCHP | + [30] | |||||
BBP | + [8], [10], [11], [12], [13], [14, [29], [30], [31], [32, 34], 36] | − [12], 31], 37], + [33] | + [8], 30], 36] | + [8], 20], 22] | ||
DBP | + [8], 9], [11], [12], [13], [14, 31], − [30] | − [12], 31], [37], [38], [39] | − [30] | + [9] | + [8], 9], 22] | |
MBP | − [9] | + [9] | + [9] | |||
DEP | + [31], − [30] | − [31] | − [30] | |||
DHP | + [8], 12], − [30] | − [12] | − [30], + [8] | + [8] | ||
DIBP | + [8], 11] | + [8] | ||||
DINP | ± [11], − [12] | − [12] | + [22] | |||
DnOP | − [12] | − [12] | + [22] | |||
DiDP | − [12] | − [12] | + [22] | |||
MBZP | − [30] | + [30] | ||||
DAP | + [8] | |||||
DPeP | + [8] | + [8] | + [8] | |||
DiHP | + [8] | + [8] | + [8] | |||
DiHepP | + [8] | + [8] | + [8] | |||
DBzP | + [36] | + [36] |
-
“+” means has potential activity; “−” means not show activity.
Moreover, it is important to note that one phthalate may exhibit several potential activities (e.g., estrogenic/anti-estrogenic or anti-androgenic) in a complex pattern.
Phthalates and epigenetic modifications
Phthalate exposure may exert its effects through epigenetic modifications, which are heritable changes in gene expression that do not involve a DNA sequence change. Epigenetic mechanisms include histone modifications such as methylation, acetylation, ubiquitination, phosphorylation, and SUMOylating, as well as DNA methylation and the expression of noncoding RNAs such as microRNAs (miRNAs). These mechanisms can influence gene expression in most cell types, and growing evidence suggests that epigenetic changes play a crucial role in the impact of environmental chemicals on human health and disease [40].
DNA methylation
As EDCs, phthalates, may exert their effects via epigenetic mechanisms such as DNA methylation, which is a frequently studied epigenetic mechanism that has been linked to both exposure and disease status. Previous studies have explored the association between phthalate exposure and DNA methylation in various organs, including the ovary, placenta, and testis [41], [42], [43], [44], [45], [46], [47], [48], as well as across different life stages, such as during prenatal development, childhood, and adulthood, and transgenerational effects, in different species including humans, mice, and fish [49], [50], [51], [52], [53], [54], [55], [56], [57], [58].
One study focused on the uterus of CD-1 mice treated with dibutyl phthalate (DBP) and reported that DBP disrupted the activity of DNA methyltransferase (DNMT) enzymes in a dose- and time-dependent manner [47]. DNMT activity in the uterus differed significantly between the DBP-treated and vehicle groups at 20 and 30 days. At 20 days of DBP exposure, only the group treated with DBP at 100 mg/kg/day showed an increase in DNMT activity. However, at 30 days, DNMT activity was reduced in all DBP-treated groups compared with the vehicle-treated group. DNMT catalyzes the transfer of a methyl group to DNA, and these changes in DNMT activity may lead to alterations in DNA methylation levels. Thus, it is reasonable to suggest that phthalate exposure may produce epigenetic changes in the uterus, which may increase the risk of developing reproductive diseases. However, direct detection of DNA methylation was not performed in this study, and further investigations are necessary to elucidate the relationship between uterine DNA methylation and phthalate exposure.
Another in vitro study investigated the relationship between phthalate exposure and DNA hypomethylation at the promoter region of the ERɑ gene [59]. MCF-7 human breast cancer cells were treated with BBP or DBP at a concentration of 10−5 M, which led to the demethylation of ERɑ promoter-associated CpG islands. F using a yeast-based ER transcription assay, these authors found that BBP treatment induced the expression of the hERɑ gene. Aberrant DNA methylation of the promoter region of the ERɑ gene might further influence the ER mRNA expression and contribute to the pathogenesis of uterine disorders.
Although several studies have explored abnormalities of DNA methylation in uterine disorders such as leiomyoma and EMs [60], [61], [62], [63], [64], few studies have investigated whether phthalate exposure increases or decreases epigenetic modifications in the uterus. Therefore, additional research is needed to elucidate the superimposed effects of phthalates on the uterus and associated diseases.
Phthalates and miRNA
Phthalates have been linked to epigenetic modifications involving miRNAs. Although the role of epigenetics-miRNA regulatory circuits in organizing the whole – gene expression profile have been investigated [65], limited studies have examined the associations between environmental phthalate exposure and miRNA expression in the uterus and associated diseases. In a recent study, Zota et al. [66] suggested that exposure to certain phthalates may be linked to miRNAs in fibroids and that the associations between exposure and miRNAs may vary according to ethnicity. The researchers quantified the expression level of 754 miRNAs in fibroid tumor samples and measured the concentration of phthalate metabolites in spot urine collected from 45 premenopausal women who underwent fibroid treatment at an academic hospital. Monohydroxy butyl phthalate (MHBP) and mono (2-ethyl-5-hydroxyhexyl) phthalate were positively associated with the expression levels of miR-10a-5p and miR-577, respectively, and some phthalate-miRNA associations varied according to ethnicity. However, in the endometrium, no significant associations were found between phthalates and miRNAs.
Considering these findings, future research should aim to evaluate prospectively the impact of environmental phthalate exposure on alterations in miRNA expression and the development of uterine disorders.
In vitro studies of endometrial cells
Uterine tissues are composed of a variety of cell types, including epithelial, stromal, endothelial and other cells, which express steroid receptors such as the ER, AR and PR. These receptors render uterine tissue susceptible to EDCs such as phthalates. In addition to differentiated cells, the endometrium also contains a distinct population of stem cells that exhibit notable regenerative capabilities during the menstrual cycle. Therefore, investigating the effects of phthalates on these cells and elucidating the potential underlying mechanisms may yield valuable evidence for characterizing these compounds in vitro.
In vitro studies of endometrial mesenchymal stem/stromal cells
Endometrial mesenchymal stem/stromal cells (EN-MSCs) are multipotent stem cells that can be isolated and cultured in vitro. They have the potential to differentiate into various cell lineages such as adipocytes, osteocytes, chondrocytes, and myocytes. Chen et al. [67] isolated and treated EN-MSCs with or without BBP and found that BBP treatment decreased EN-MSCs differentiation. Further evidence suggested that BBP decreased myogenic differentiation of EN-MSC via miR-137-mediated regulation of PITX2. These findings suggest that exposure to phthalates may inhibit the differentiation of EN-MSCs, which may affect endometrial regeneration. However, the precise mechanism requires further exploration.
In vitro study of endometrial endothelial cells
The endometrium is regulated by steroid hormones and undergoes regeneration during each menstrual cycle. A vital component of the endometrium, vascular trees play a crucial role in the control of bleeding and fertility. Endothelial cells lining the luminal surface of all blood vessels modulate their vascular morphology and function through angiogenesis, vascular remodeling, and functional changes. Human endometrial endothelial cells (HEECs) are unique in their expression of the ERβ and PR. Dysfunction of these cells is associated with endometrial disorders including EMs and abnormal uterine bleeding [68], 69].
As previously mentioned, some phthalates exhibit potential estrogenic activity. Because HEECs express the relevant acceptors, it is imperative to investigate whether these compounds affect cell function. Bredhult et al. [70] reported that exposure to DBP decreased the proliferation of in vitro-cultured HEECs; however, the underlying mechanism remains to be elucidated. In a recent study, Lijuan An [71] observed that exposure to mono (2-ethylhexyl) phthalate (MEHP) inhibited the proliferation of human endometrial microvascular endothelial cells (HEMECs) and induced apoptosis and pyroptosis via NLRP3 inflammasome. Given the widespread use of phthalates and the diversity of these compounds, further investigations are needed to explore the effects of other phthalates on HEECs.
In vitro studies of endometrial stromal cells
Endometrial stromal cells (ESCs) are an important cell type in the female reproductive system and their response to phthalates has been studied in vitro [72]. In 2010, Kim et al. cultured ESC under serum-free conditions and exposed them to hydrogen peroxide. They observed for the first time that di-(2-ethylhexyl) phthalate (DEHP) exposure led to increased ESC viability. Additional research confirmed Kim’s these results and demonstrated that DEHP exposure increased the production of reactive oxygen species (ROS) and decreased the expression of glutathione peroxidase (GPX), superoxide dismutase (SOD), catalase (CAT), and heme oxygenase (HO) in human ESCs [73]. In that study, DEHP exposure induced the expression of the ERα, which phosphorylated extracellular signal-regulated kinase (p-ERK)/p-p38 MAPK and NF-κB pathways and mediated the transcription of downstream genes. Another recent study found that DEHP treatment increased ESC proliferation and migration, inflammatory responses, and induction of the epithelial-mesenchymal transition (EMT) and stemness in human endometrial and endometriotic cells through targeting the TGF-β/Smad signaling pathway [74]. In contrast to the data reported by Kim et al. [72], other studies [75], [76], [77] have reported no significant effects on ESC viability following DEHP exposure. These discrepancies may reflect differences in treatment conditions or data analysis methods. For instance, the addition of hydrogen peroxide in the study by Kim et al. may have influenced cell viability.
Other research has investigated the impact of phthalates on the viability of ESCs, and some have shown that treatment with DEHP in in vitro-cultured endometrial cells significantly increased cellular invasiveness [77], activities of matrix metalloproteinase-2 (MMP-2) and 9 [78], 79], phosphorylation of extracellular signal-regulated kinase (Erk), and expression of p21-activated kinase 4 (Pak4) [80]. Further, increased expression of inflammation factors (IL-1β, IL-8, ICAM-1, and COX-2), peroxisome proliferator-activated receptor gamma (PPARγ), and aldose-keto reductase (AKR) activity (AKR1C1-3 and AKR1B10) was also observed [75], 81], 82]. All of the aforementioned genes may contribute to various gynecological disorders, such as EMs and endometrial cancer.
In addition to the aforementioned studies, one study [76] of cows has shown that DEHP and MEHP treatment of cultured endometrial cells stimulated the production of prostaglandin F2α (PGF2α) and inhibited the secretion of PGE2α, thereby increasing the PGF2α:PGE2α ratio. Such alterations may result in increased myometrial contractions and accelerate luteal regression, which cold lead to dysmenorrhea and abnormal uterine bleeding. Recent studies have found that DEHP can induce endometrial decidualization defects, which may affect embryo implantation and development, and lead to female reproductive disorders [83], 84].
Overall, the data suggests that exposure to phthalates such as DEHP or MEHP may alter the viability of endometrial cells and change their gene expression patterns (as shown in Figure 1). Such exposure may increase the expression of ERα, increase ROS production, and activate the MAPK/ERK/PAK4 and NF-κB pathways, which may further regulate the expression of genes associated with uterine disorders such as ER, PR, E-cadherin, AKR1C1-3, AKR1B10, MMP2/9.

Potential mechanism(s) of the role of phthalates in uterine disorders. In the “genomic pathway”, many actions of phthalates are mediated by the classical nuclear hormone receptors (NRs), especially estrogen receptors (ERs) and progesterone receptors (PR). After binding to NRs, phthalates can affect the transcription of target genes in the nucleus by binding to the corresponding response element of target genes. The “non-genomic pathway” of phthalates may occur through membrane steroid receptors (ERs, PRs), which may lead to rapid downstream intracellular signaling (MAPK/ERK) or through tryptophan-kynurenine – aryl hydrocarbon receptor (AHR) pathway and regulate the gene transcription. The “epigenetic modification” of phthalates may occur through DNA methylation and then regulate target gene expression.
Phthalate and uterine leiomyoma (fibroid)
Uterine leiomyoma is the most frequent benign gynecologic tumor, and 70–80 % of women may be affected by this condition over their lifetime. The presence of uterine leiomyoma can affect a women’s quality of life and may require surgery such as myomectomy or hysterectomy. Many risk factors for the development of this tumor including phthalates exposure have been identified. In a comprehensive review of the literature on the effects of phthalates on uterine leiomyoma. We identified 14 epidemiological studies [85], [86], [87], [88], [89], [90], [91], [92], [93], [94], [95], [96], [97], [98] and one meta-analysis [99], of these studies, only one measured serum phthalate metabolites while the other 13 studies (shown in Table 3) used urine samples. However, the results of these studies were inconsistent.
Evidence profile table of studies of the associations between phthalate exposure and leiomyoma.
Study, year | Study design | Time period | Age range | Study population | No. of case/control | Samples | Categories of phthalates and metabolites | Summary of findings |
---|---|---|---|---|---|---|---|---|
Luisi et al. 2006 [89] | Case-control | Not mentioned | Case: 48–50 | General population | 15/20 | Serum | DEHP MEHP | Serum DEHP and MEHP concentration were lower in women with uterine fibromatosis |
Control: not mentioned | ||||||||
Huang et al. 2010 [86] | Case-control | 2005–2007 | 27–45 (41.1 ± 6.8) |
Laparotomy population | 36/29 | Urine | MMP, MEP, MnBP, MBzP, 5oxo-MEHP, 5OH-MEHP, MEHP | MMP, MEHP and ΣMEHP were significantly higher in the LEI patients than in controls. |
Weuve et al. 2010 [92] | Cross-sectional | 1999–2004 | 20–54 | General population | 151/1,020 | Urine | MBP, MEP, MEHP, MBzP, MEHHP, MEOHP | Positive association for MBP and inverse association for MEHP in relation to leiomyoma. |
Huang et al. 2014 [85] | Case-control | 2005–2007 | 41.1 ± 6.8 | Laparotomy population | 36/69 | Urine | MMP, MEP, MnBP, MBzP, 5oxo-MEHP, 5OH-MEHP, MEHP | Compared with controls, patients with leiomyoma had significantly higher levels of total urinary ΣMEHP, MnBP, and MEP. |
Pollack et al. 2015 [90] | Case-control | 2007–2009 | Case: 37.8 ± 4.9 | Laparotomy population | 99/374 | Urine | MBzP, MnBP, MiBP, MCHP, MEP, MEHP, MEHHP, MEOHP, MECPP, MiNP, MCNP, MMP, MCPP, MOP | MMP levels were significantly lower in women with than without fibroids. |
Control: 31.7 ± 6.9 | ||||||||
Kim et al. 2016 [88] | Case-control | March 2013 and July 2015 | Case: 42.6 ± 1.02 | Laparotomy population | 30/27 | Urine | MBzP, MiBP, MCHP, MEP, MEHP, MEHHP, MEOHP, MECPP, 2cx-MMHP 5cx-MEPP, MMP, MCPP, MnOP, MnPP, MiDP, MiNP | The log transformed creatinine-adjusted levels of MEHP, MEHHP, 2cx-MMHP and ΣDEHP were significantly higher in the leiomyoma group than in the controls. |
Con: 34.78 ± 1.90; | ||||||||
Sun et al. 2016 [91] | Case-control | 2010.10–2012.05 | Reproductive age | General population | 61/61 | Urine | MMP, MEP, MiBP, MnBP, MEHP, MEHHP, MEOHP, MECPP, MCMHP | Cases had significantly higher levels of creatinine-adjusted MiBP, MnBP, MEHP, MEOHP, MEHHP, MECPP, total ΣDEHPmet, and total ΣDBPmet than controls. |
Kim et al. 2017 [87] | Case-control | March 2012 and February 2013 | Case: 35.3 ± 0.8 | Surgery population | 53/33 | Urine | MEHHP, MEOHP, MnBP, MBzP, MECPP | The urinary concentration of MECPP was higher in women with leiomyoma than in controls. |
Con: 32.6 ± 1.41; | ||||||||
Zota et al. 2019 [98] | Pilot, cross-sectional study | During 2014–2017 | 26–54 | Women with symptomatic fibroid tumors and subsequently undergoing surgical management | 57 | Urine | MEP, MnBP, MHBP, MiBP, MHiBP, MBzP, MCPP, MiNP, MCOP, MCNP, MEHP, MEHHP, MEOHP, and MECPP and ΣDEHP | Higher levels of MHiBP, MCOP, MCNP, MEHHP, MEOHP, and MECPP and ΣDEHP phthalates were positively associated with uterine volume. |
Lee et al. 2020 [93] | Case-control | During 2015–2016 | Case: 38.2 ± 5.3 | General population | 32/79 | Urine | OH-MINP, cxMINP, MMP, MEP, MiPP, MiBP, MBP, MPeP, MBzP, MCHP, MHxP, MEOHP, MEHHP, MECPP, MCMHP, MEHP, ΣDEHP metabolites, MCPP | The concentrations of MEOHP, ΣDEHP, and OH-MINP in urine were significantly higher in the cases than in the control group. |
Con: 37.8 ± 5.11 | ||||||||
Lee et al. 2020 [94] | Case-control | During 2015–2016 | 25–45 | General population | 96/337 | Urine | MMP, MEP, MiBP, MnBP, MBzP, MECPP, MCMHP, MEHHP, MEOHP, ΣDEHP metabolites | Urinary concentration of MMP, MECPP, ΣDEHP metabolites were associated with increased odds of leiomyoma. |
Fruh et al. 2021 [95] | An ongoing prospective cohort study | 2010–2012 | A prospective cohort of premenopausal Black women aged 23–35 years | A population of reproductive-aged Black women | 754 Black women | Urine | MBP, MHBP, MiBP, MHiBP, MBzP, MEHHP, MEHP, MEOHP, MECPP, MEP, MCOP, MNP, MCNP, MCPP. | The urinary concentrations of phthalate biomarkers were not significantly associated with higher risk of uterine leiomyoma, either individually or jointly. |
Pacyga et al. 2022 [96] | A retrospective study | Women from NHANES 2005–2016 and MWHS 2006–2015 | Ages: 45–54 years | Pre- and peri-menopausal women | 754 | Urine | MEHP, MEHHP, MECPP, MEOHP, MCPP, MBzP, MEP, MBP, MiBP ΣDEHP | The risk of prior fibroid diagnosis was increased by 13 % for each two-fold increase in ΣDEHP. |
Zhang et al. 2023 [97] | Case-control | 2018–2020 | Case: 30.7 ± 4.3 | Females who went through ART treatment | 226/83 | Urine | MEP, MiBP, MBP, MBzP, MEHP, MEOHP, MEHHP, MECPP | Increased urinary MBzP concentration was associated with an increased risk of uterine fibroids. |
Con: 33.6 ± 5.0 |
-
Phthalates metabolites with a negative association with fibroids are shown in gray, those with a positive association are shown in bold letters, and those with no significant difference are shown in black normal font letter. MWHS, the Midlife Women’s Health Study; NHANES, the National Health and Nutrition Examination Survey.
In one study by Luisi et al. [89], DEHP and MEHP concentrations were lower in women with uterine fibromatosis. Another study that tested urine samples also found an inverse association between MEHP and leiomyoma [92]. In a separate study, the level of one common phthalate metabolites (MMP) in urine was significantly lower in women with than in those without fibroids (1.78 vs. 2.40 mg/g, respectively) [90]. However, other studies have found that patients with leiomyoma had significantly higher levels of at least one urine phthalates metabolites (as shown in bold in Table 3) compared with control [[85], [86], [87], [88, 91], [93], [94], [95, 97]. A Pilot, cross-sectional study found a positive association between phthalate exposure and uterine volume [98]. Despite conflicting findings, the 2017 meta-analysis by Fu et al. found a significant positive association between DEHP metabolites and uterine leiomyoma [99]. In a study by Pacyga et al. [96], women had a 13 % greater risk of prior fibroid diagnosis for a twofold increase in ΣDEHP (MEHP, MEHHP, MEOHP, and MECPP), which suggested a positive relationship between the presence of fibroids and DEHP. A prospective ultrasound study investigated the association between phthalate metabolites and uterine leiomyoma incidence, and found that MEHP concentration was positively associated with the incidence of uterine leiomyoma, whereas other metabolites had an inverse relationship. However, these associations were weak to moderate and did not reach statistical significance [95]. All of these results suggest that DEHP and its metabolites are positively associated with the development of fibroids.
DEHP is an EDC that is associated with several adverse health outcomes, including adverse reproductive outcomes in both men and women, insulin resistance, type II diabetes, overweight/obesity, asthma, and allergy [100]. In vitro studies [87], 101] have suggested that DEHP may play a role in the pathogenesis of uterine leiomyoma by exerting an antiapoptotic effect, increasing proliferative activity, and increasing collagen production in myometrial and leiomyoma cells. A more recent in vitro study revealed that the principal DEHP metabolite, MEHHP, promotes leiomyoma cell survival by activating the tryptophan-kynurenine-aryl hydrocarbon receptor pathway [102]. A further in vivo study found that NOD/SCID mice implanted with fibroid tissues and fed DEHP had larger volumes of fibroid tissues than control mice that were not exposed to DEHP [103]. Therefore, it is reasonable to propose that this phthalate may have a negative role in uterine leiomyoma. In another study, exposure to certain phthalate biomarkers, such as mono-hydroxyisobutyl phthalate (MHiBP), monocarboxyoctyl phthalate (MCOP), monocarboxynonyl phthalate (MCNP), MEHP, MEHHP, MEOHP, and MECPP, was positively associated with the volume of the uterus; this finding provides further evidence that phthalate exposure may be associated with fibroid outcomes [98].
Phthalate and endometriosis/adenomyosis
EMs is defined as the presence of endometrial-type mucosa outside the uterine cavity. When endometrial tissues invade the myometrium with smooth muscle hyperplasia, it is defined as adenomyosis (AD). EMs is one of the most common benign gynecological diseases in premenopausal women with morbidity 30–50 % in women with infertility and 10–15 % of reproductive-aged women. Patients diagnosed with EMs commonly present with clinical symptoms such as pelvic pain, adnexal masses, or infertility. These patients may also have an increased risk of epithelial ovarian cancer [104], 105]. Endometriosis-associated ovarian cancers, which include endometrioid and clear cell ovarian carcinoma, are generally acknowledged as the most lethal gynecological malignancy [104]. Although the potential pathogenesis of EMs and AD remains unclear, environmental factors such as phthalates are likely to play important roles.
Using a biomimetic hydrogel-based three-dimensional culture model, Kim et al. [74] suggested that DEHP could increase the proliferation and migration of human endometrial and endometriotic epithelial cells, induce the EMT, and promote stem cell properties via the TGF-β/Smad signaling pathway,. This research provides a new potential therapeutic target for DEHP-induced endometriosis.
According to in vitro studies, phthalates exposure may increase the proliferation and viability of endometrial cells, which may lead to the development of EMs via retrograde menstruation. In an in vivo study using the NOD/SCID mouse model, Kim et al. found that the size of endometrial implants was significantly larger in mice fed DEHP compared with those fed with vehicle alone. Although this model cannot fully replicate the human immune system, the findings support the idea that DEHP may stimulate the formation of EMs. A study by Sharma et al. reported that chronic exposure to low-dose BBP promote survival of endometriotic tissue through CD44-expressing on plasmacytoid dendritic cells [106]. In this study, BBP exposure did not promote the growth of endometriotic lesions, although the lesion survival rate in BBP-treated mice was significantly higher than in the control mice.
We conducted a search of all relevant literature for human until Nov 31, 2023 and identified 17 original studies that investigated the relationship between phthalate exposure and EMs (16 studies) [81], 85], 86], 92], 97], [107], [108], [109], [110], [111], [112], [113], [114], [115], [116], [117] or AD (2 studies) [86], 94] which are summarized in Table S1. However, the conclusions of these studies are inconsistent. As shown in Table 4, some studies reported results that are completely opposite, such as those involving DEHP and its metabolites. For example, Cobellis et al. [107], Reddy et al. [109], Kim et al. [111], Nazir et al. [114], and Yi et al. [117] found that women with EMS had significantly higher plasma DEHP concentration than the controls, which suggested a positively association between DEHP concentration and EMs. Upson et al. [113] reported a decreasing trend in the EMs group despite the lack of significant difference in concentration between the Ems and the control group. These differences may reflect differences in samples used (urine and plasma) and study populations. Other studies have also found that the concentrations of MEHP and DHEP are associated with EMs and with advanced-stage EMs [107], 108].
Associations between phthalates and EMs in 16 studies. Level of evidence of phthalate exposure related to EMs was highlighted according to an adapted version of the Oxford Center for Evidence-Based Medicine (CEBM) and Marx et al. [121], [122], [123].
![]() ![]() ![]() |
Contradictory results were found more often for DEHP and its primary or secondary metabolites. Phthalate diesters are unstable in human blood and have a short half-life (e.g., 28 min for DEHP). Serum enzymes are reported to hydrolyze DEHP to MEHP during storage [118], 119], and blood sampling can cause phthalate exposure. Given these factors, blood phthalate diesters may be less appropriate biomarkers for evaluating phthalate exposure. Although the results for the metabolites of DEHP in urine were also inconsistent, urinary phthalate metabolites are more stable and relatively free from laboratory contamination.
The evidence supporting a relationship between MBP (MnBP) and EMS is also inconsistent. Five studies [86], 92], 112], 116], 117] have suggested a positive relationship between MBP and EMs, but six other studies [81], 85], 97], 110], 113], 115] have shown no significant difference between the EMs and control groups. Other phthalates such as BBzP, MMP, MiBP, MEP, MBzP also have contradictory effects as shown in Table 4. The discrepant findings reflect differences in exposure assessment and/or case definition. Among these studies, two studies from the same research group have made different conclusions, which may have been influenced by the large standard deviations and individual variability. However, the trend for MBP is the same, with higher concentrations found in the EMs group than in the control groups [85], 92]. No associations have been found between EMs and other phthalates, including MOP, MCHP, and MINP, which suggests that these phthalates may have weak estrogenic activity.
In a meta-analysis, Cai et al. [120] investigated five types of phthalate metabolites (MEHHP, MEP, MBzP, MEHP, and MEOHP) and found that only one metabolite (MEHHP) was potentially associated with EMs. However, this meta-analysis included only eight studies, the quality of the studies was moderate, the available evidence was limited, and there may have been some bias.
No association was found between the exposure to phthalates and AD [86], 94]. However, given that there are only two studies, and their samples were small, further investigation is needed.
In conclusion, given that EMs is an estrogenic-associated disease and phthalates have potential estrogenic activity, there appears to be a strong association between phthalate exposure and this disease. However, further studies are needed to clarify this relationship. Future research should include larger sample sizes and more phthalate metabolites. Additionally, the sample detection time and methods should be taken into consideration to improve the accuracy and comparability of the results.
Phthalates and uterine histology changes
Phthalates exhibit estrogenic activity and can regulate endometrial cells in vitro. In recent years, the effects of phthalate exposure on the uterus have been explored in vivo.
Exposure to phthalates, such as DEHP and DEP, may affect uterus weight, although findings are not always consistent. Some studies have found that DEHP exposure increased uterus weight, but others have reported no significant change, and one study found a decrease in weight in immature rats exposed to DEHP during postnatal day 1–21 [124], [125], [126], [127], [128]. These discrepancies may reflect differences in exposure conditions, such as the time, dose, duration, and age of exposure. DEP, another common phthalate, has also been shown to increase the height of uterine epithelial cells as well as uterine weight in adult female rats. Moreover, DEP exposure up-regulates the expression of several steroidogenic genes and the pS2 gene and activates the MAPK pathway by increasing the p-ERK/ERK ratio [129].
Somasundaram et al. [126] demonstrated that exposing adult rats to DEHP for 30 days at doses of 0–100 mg/kg body weight/day increased the protein levels of ERs, PRs and PPARγ in the uterus. However, no significant changes were observed in the wet uterine weight. Histological studies revealed a decreased diameter and layers of the uterus and disrupted glandular epithelium. These findings are inconsistent with those of Richardson et al. [124], who found that DEHP exposure for 30 days at different doses (20 ug/kg/day to 200 mg/kg/day) increased the number of uterine glands, increased the proliferation of uterine stromal cells at the highest dose, and increased the numbers of weakened, dilated blood vessels in the endometrium. Kim et al. [125] found that chronic low-dose exposure to DEHP had an estrogen-like effect in the mouse uterus. In contrast to the data of Somasundaram et al. [126] but consistent with those of Richardson et al. [124], Kim et al. found that DEHP exposure increased the ratio of uterine weight to body weight (133 μg/L), endometrial and myometrial thickness (133 and 1,330 μg/L), and numbers of luminal epithelial cells (50 μg/L). In addition, the mRNA expression levels of known 17β-estradiol (E2) downstream genes, Esr1, Esr2, Prg, Lox, Egr1 and Muc1 were also changed. Further experiments found that DEHP changed the proliferation status of uterine endometrial cells and modified the translocation of ESR1, ESR2 and PGR [130]. These results suggest that DEHP affects the nuclear activation of steroid hormonal receptors, which may induce histological changes in a dose-dependent manner and have adverse effects on normal uterus function.
DEHP exposure not only has effects on the mother’s generation (F0), but also has long-term reproductive toxic effects on the offspring, F1 and F2, such as expression of the PR, ERα. The expression of estrogen and progesterone regulatory genes such as HoxA11, VEGF A, IHH, LIFR, EP4, PTCH, NR2F2, BMP2, and WNT4 in the uterus were reported to be decreased in DEHP-exposed offspring rats [131]. In another study, female offspring of female mice exposed to DEHP from prenatal to peripubertal times developed endometrial atrophy and fibrosis [132].
Li et al. [133] found that prenatal exposure to a mixture of phthalates in mice (including 5 % BBP, 8 % DIBP, 15 % DINP, 15 % DBP, 21 % DEHP, and 35 % DEP) led to transgenerational and multigenerational effects on uterine morphology and function. The exposure to the mixture (20 μg/kg/day to 500 mg/kg/day) from gestational day 10.5 to parturition resulted in a higher incidence of large, dilated glands, multilayered luminal epithelium, and fibrotic response in all generations. The luminal epithelial cell proliferation was also increased in F2 generation in the 200 mg/kg/day group. A recent study also found that chronic exposure to a phthalate mixture increased the thickness of the myometrial layer and the incidence of collagen deposition in uteri, which may be associated with the upregulation of TGF-β signaling [134]. Although some studies have mentioned the effects of phthalate exposure on uterus weight and cell proliferation, few have explored the underlying mechanisms. Moreover, some studies have shown that prenatal exposure to phthalates such as DiNP has no influence on uterus weight [135], 136]. Further research is needed to examine the effects of phthalate exposure on endometrial hyperplasia and oncogenesis, as well as on the cellular function of the uterus.
Conclusions
In conclusion, studies conducted both in vitro, in vivo and on human populations suggest a strong association between exposure to phthalates and uterus disorders, such as leiomyoma and EMs. Figure 1 summarizes the potential mechanisms underlying this association. However, whether phthalates play a role in uterine carcinomas require further investigation. Abnormal uterine bleeding which is associated with hormone fluctuation and the endometrial environment, including inflammatory factors, may be affected by phthalate exposure. The understanding of the effects of phthalates on the endometrial environment and its secretory functions is currently limited. Research into the epigenetic changes in the uterus caused by phthalate exposure may reveal new insights into the effects of EDCs. Therefore, exploring the effects of phthalate exposure and its underlying mechanisms is crucial for discovering new interventions to prevent and treat uterine disorders in women.
-
Research ethics: Not applicable.
-
Informed consent: Not applicable.
-
Author contributions: The first draft of the manuscript was written by Shuhong Yang. Shuhao Yang and Aiyue Luo revised the review. All authors read and approved the final manuscript.
-
Competing interests: The authors declare no competing interests.
-
Research funding: Not applicable.
-
Data availability: Not applicable.
References
1. Huang, PC, Chang, WH, Wu, MT, Chen, ML, Wang, IJ, Shih, SF, et al.. Characterization of phthalate exposure in relation to serum thyroid and growth hormones, and estimated daily intake levels in children exposed to phthalate-tainted products: a longitudinal cohort study. Environ Pollut 2020;264:114648. https://doi.org/10.1016/j.envpol.2020.114648.Search in Google Scholar PubMed
2. Wang, Y, Zhu, H, Kannan, K. A review of biomonitoring of phthalate exposures. Toxics 2019;7:21. https://doi.org/10.3390/toxics7020021.Search in Google Scholar PubMed PubMed Central
3. Eales, J, Bethel, A, Galloway, T, Hopkinson, P, Morrissey, K, Short, RE, et al.. Human health impacts of exposure to phthalate plasticizers: an overview of reviews. Environ Int 2022;158:106903. https://doi.org/10.1016/j.envint.2021.106903.Search in Google Scholar PubMed
4. Ashworth, MJ, Chappell, A, Ashmore, E, Fowles, J. Analysis and assessment of exposure to selected phthalates found in children’s toys in Christchurch, New Zealand. Int J Environ Res Public Health 2018;15:200. https://doi.org/10.3390/ijerph15020200.Search in Google Scholar PubMed PubMed Central
5. Mertens, HJ, Heineman, MJ, Theunissen, PH, de Jong, FH, Evers, JL. Androgen, estrogen and progesterone receptor expression in the human uterus during the menstrual cycle. Eur J Obstet Gynecol Reprod Biol 2001;98:58–65. https://doi.org/10.1016/s0301-2115(00)00554-6.Search in Google Scholar PubMed
6. Begum, TF, Carpenter, D. Health effects associated with phthalate activity on nuclear receptors. Rev Environ Health 2022;37:567–83. https://doi.org/10.1515/reveh-2020-0162.Search in Google Scholar PubMed
7. Alva-Gallegos, R, Carazo, A, Mladenka, P. Toxicity overview of endocrine disrupting chemicals interacting in vitro with the oestrogen receptor. Environ Toxicol Pharmacol 2023;99:104089. https://doi.org/10.1016/j.etap.2023.104089.Search in Google Scholar PubMed
8. Takeuchi, S, Iida, M, Kobayashi, S, Jin, K, Matsuda, T, Kojima, H. Differential effects of phthalate esters on transcriptional activities via human estrogen receptors alpha and beta, and androgen receptor. Toxicology 2005;210:223–33. https://doi.org/10.1016/j.tox.2005.02.002.Search in Google Scholar PubMed
9. Shen, O, Du, G, Sun, H, Wu, W, Jiang, Y, Song, L, et al.. Comparison of in vitro hormone activities of selected phthalates using reporter gene assays. Toxicol Lett 2009;191:9–14. https://doi.org/10.1016/j.toxlet.2009.07.019.Search in Google Scholar PubMed
10. Andersen, HR, Andersson, AM, Arnold, SF, Autrup, H, Barfoed, M, Beresford, NA, et al.. Comparison of short-term estrogenicity tests for identification of hormone-disrupting chemicals. Environ Health Perspect 1999;107:89–108. https://doi.org/10.1289/ehp.99107s189.Search in Google Scholar PubMed PubMed Central
11. Harris, CA, Henttu, P, Parker, MG, Sumpter, JP. The estrogenic activity of phthalate esters in vitro. Environ Health Perspect 1997;105:802–11. https://doi.org/10.2307/3433697.Search in Google Scholar
12. Zacharewski, TR, Meek, MD, Clemons, JH, Wu, ZF, Fielden, MR, Matthews, JB. Examination of the in vitro and in vivo estrogenic activities of eight commercial phthalate esters. Toxicol Sci 1998;46:282–93. https://doi.org/10.1093/toxsci/46.2.282.Search in Google Scholar
13. Jobling, S, Reynolds, T, White, R, Parker, MG, Sumpter, JP. A variety of environmentally persistent chemicals, including some phthalate plasticizers, are weakly estrogenic. Environ Health Perspect 1995;103:582–7. https://doi.org/10.2307/3432434.Search in Google Scholar
14. Ghisari, M, Bonefeld-Jorgensen, EC. Effects of plasticizers and their mixtures on estrogen receptor and thyroid hormone functions. Toxicol Lett 2009;189:67–77. https://doi.org/10.1016/j.toxlet.2009.05.004.Search in Google Scholar PubMed
15. Nomura, Y, Mitsui, N, Bhawal, UK, Sawajiri, M, Tooi, O, Takahashi, T, et al.. Estrogenic activity of phthalate esters by in vitro VTG assay using primary-cultured Xenopus hepatocytes. Dent Mater J 2006;25:533–7. https://doi.org/10.4012/dmj.25.533.Search in Google Scholar PubMed
16. Yamasaki, K, Takeyoshi, M, Sawaki, M, Imatanaka, N, Shinoda, K, Takatsuki, M. Immature rat uterotrophic assay of 18 chemicals and Hershberger assay of 30 chemicals. Toxicology 2003;183:93–115. https://doi.org/10.1016/s0300-483x(02)00445-6.Search in Google Scholar PubMed
17. Ter Veld, MG, Zawadzka, E, van den Berg, JH, van der Saag, PT, Rietjens, IM, Murk, AJ. Food-associated estrogenic compounds induce estrogen receptor-mediated luciferase gene expression in transgenic male mice. Chem Biol Interact 2008;174:126–33. https://doi.org/10.1016/j.cbi.2008.03.019.Search in Google Scholar PubMed
18. Yamasaki, K, Sawaki, M, Noda, S, Muroi, T, Takakura, S, Mitoma, H, et al.. Comparison of the Hershberger assay and androgen receptor binding assay of twelve chemicals. Toxicology 2004;195:177–86. https://doi.org/10.1016/j.tox.2003.09.012.Search in Google Scholar PubMed
19. Araki, N, Ohno, K, Nakai, M, Takeyoshi, M, Iida, M. Screening for androgen receptor activities in 253 industrial chemicals by in vitro reporter gene assays using AR-EcoScreen cells. Toxicol Vitro 2005;19:831–42. https://doi.org/10.1016/j.tiv.2005.04.009.Search in Google Scholar PubMed
20. Kruger, T, Long, M, Bonefeld-Jorgensen, EC. Plastic components affect the activation of the aryl hydrocarbon and the androgen receptor. Toxicology 2008;246:112–23. https://doi.org/10.1016/j.tox.2007.12.028.Search in Google Scholar PubMed
21. Roy, P, Salminen, H, Koskimies, P, Simola, J, Smeds, A, Saukko, P, et al.. Screening of some anti-androgenic endocrine disruptors using a recombinant cell-based in vitro bioassay. J Steroid Biochem Mol Biol 2004;88:157–66. https://doi.org/10.1016/j.jsbmb.2003.11.005.Search in Google Scholar PubMed
22. Lee, BM, Koo, HJ. Hershberger assay for antiandrogenic effects of phthalates. J Toxicol Environ Health Part A 2007;70:1365–70. https://doi.org/10.1080/15287390701432285.Search in Google Scholar PubMed
23. Richard, AM, Judson, RS, Houck, KA, Grulke, CM, Volarath, P, Thillainadarajah, I, et al.. ToxCast chemical landscape: paving the road to 21st century toxicology. Chem Res Toxicol 2016;29:1225–51. https://doi.org/10.1021/acs.chemrestox.6b00135.Search in Google Scholar PubMed
24. Judson, RS, Magpantay, FM, Chickarmane, V, Haskell, C, Tania, N, Taylor, J, et al.. Integrated model of chemical perturbations of a biological pathway using 18 in vitro high-throughput screening assays for the estrogen receptor. Toxicol Sci 2015;148:137–54. https://doi.org/10.1093/toxsci/kfv168.Search in Google Scholar PubMed PubMed Central
25. Kleinstreuer, NC, Ceger, P, Watt, ED, Martin, M, Houck, K, Browne, P, et al.. Development and validation of a computational model for androgen receptor activity. Chem Res Toxicol 2017;30:946–64. https://doi.org/10.1021/acs.chemrestox.6b00347.Search in Google Scholar PubMed PubMed Central
26. Ruiz, P, Sack, A, Wampole, M, Bobst, S, Vracko, M. Integration of in silico methods and computational systems biology to explore endocrine-disrupting chemical binding with nuclear hormone receptors. Chemosphere 2017;178:99–109. https://doi.org/10.1016/j.chemosphere.2017.03.026.Search in Google Scholar PubMed PubMed Central
27. Manganelli, S, Roncaglioni, A, Mansouri, K, Judson, RS, Benfenati, E, Manganaro, A, et al.. Development, validation and integration of in silico models to identify androgen active chemicals. Chemosphere 2019;220:204–15. https://doi.org/10.1016/j.chemosphere.2018.12.131.Search in Google Scholar PubMed PubMed Central
28. Najjar, A, Wilm, A, Meinhardt, J, Mueller, N, Boettcher, M, Ebmeyer, J, et al.. Evaluation of new alternative methods for the identification of estrogenic, androgenic and steroidogenic effects: a comparative in vitro/in silico study. Arch Toxicol 2024;98:251–66. https://doi.org/10.1007/s00204-023-03616-y.Search in Google Scholar PubMed PubMed Central
29. Picard, K, Lhuguenot, JC, Lavier-Canivenc, MC, Chagnon, MC. Estrogenic activity and metabolism of n-butyl benzyl phthalate in vitro: identification of the active molecule(s). Toxicol Appl Pharmacol 2001;172:108–18. https://doi.org/10.1006/taap.2001.9141.Search in Google Scholar PubMed
30. 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;26:1219–24. https://doi.org/10.1248/bpb.26.1219.Search in Google Scholar PubMed
31. Hong, EJ, Ji, YK, Choi, KC, Manabe, N, Jeung, EB. Conflict of estrogenic activity by various phthalates between in vitro and in vivo models related to the expression of Calbindin-D9k. J Reprod Dev 2005;51:253–63. https://doi.org/10.1262/jrd.16075.Search in Google Scholar PubMed
32. Fujita, T, Kobayashi, Y, Wada, O, Tateishi, Y, Kitada, L, Yamamoto, Y, et al.. Full activation of estrogen receptor alpha activation function-1 induces proliferation of breast cancer cells. J Biol Chem 2003;278:26704–14. https://doi.org/10.1074/jbc.m301031200.Search in Google Scholar PubMed
33. Laws, SC, Carey, SA, Ferrell, JM, Bodman, GJ, Cooper, RL. Estrogenic activity of octylphenol, nonylphenol, bisphenol A and methoxychlor in rats. Toxicol Sci 2000;54:154–67. https://doi.org/10.1093/toxsci/54.1.154.Search in Google Scholar PubMed
34. Park, C, Lee, J, Kong, B, Park, J, Song, H, Choi, K, et al.. The effects of bisphenol A, benzyl butyl phthalate, and di(2-ethylhexyl) phthalate on estrogen receptor alpha in estrogen receptor-positive cells under hypoxia. Environ Pollut 2019;248:774–81. https://doi.org/10.1016/j.envpol.2019.02.069.Search in Google Scholar PubMed
35. Stroheker, T, Cabaton, N, Nourdin, G, Regnier, JF, Lhuguenot, JC, Chagnon, MC. Evaluation of anti-androgenic activity of di-(2-ethylhexyl)phthalate. Toxicology 2005;208:115–21. https://doi.org/10.1016/j.tox.2004.11.013.Search in Google Scholar PubMed
36. Zhang, Z, Hu, Y, Zhao, L, Li, J, Bai, H, Zhu, D, et al.. Estrogen agonist/antagonist properties of dibenzyl phthalate (DBzP) based on in vitro and in vivo assays. Toxicol Lett 2011;207:7–11. https://doi.org/10.1016/j.toxlet.2011.08.017.Search in Google Scholar PubMed
37. Ahmad, R, Verma, Y, Gautam, AK, Kumar, S. Assessment of estrogenic potential of di-n-butyl phthalate and butyl benzyl phthalate in vivo. Toxicol Ind Health 2015;31:1296–303. https://doi.org/10.1177/0748233713491803.Search in Google Scholar PubMed
38. Kim, HS, Kang, TS, Kang, IH, Kim, TS, Moon, HJ, Kim, IY, et al.. Validation study of OECD rodent uterotrophic assay for the assessment of estrogenic activity in Sprague-Dawley immature female rats. J Toxicol Environ Health Part A 2005;68:2249–62. https://doi.org/10.1080/15287390500182354.Search in Google Scholar PubMed
39. Papaconstantinou, AD, Fisher, BR, Umbreit, TH, Brown, KM. Increases in mouse uterine heat shock protein levels are a sensitive and specific response to uterotrophic agents. Environ Health Perspect 2002;110:1207–12. https://doi.org/10.1289/ehp.021101207.Search in Google Scholar PubMed PubMed Central
40. Cavalli, G, Heard, E. Advances in epigenetics link genetics to the environment and disease. Nature 2019;571:489–99. https://doi.org/10.1038/s41586-019-1411-0.Search in Google Scholar PubMed
41. Rattan, S, Beers, HK, Kannan, A, Ramakrishnan, A, Brehm, E, Bagchi, I, et al.. Prenatal and ancestral exposure to di(2-ethylhexyl) phthalate alters gene expression and DNA methylation in mouse ovaries. Toxicol Appl Pharmacol 2019;379:114629. https://doi.org/10.1016/j.taap.2019.114629.Search in Google Scholar PubMed PubMed Central
42. Strakovsky, RS, Schantz, SL. Impacts of bisphenol A (BPA) and phthalate exposures on epigenetic outcomes in the human placenta. Environ Epigenet 2018;4:dvy022. https://doi.org/10.1093/eep/dvy022.Search in Google Scholar PubMed PubMed Central
43. Zhao, Y, Chen, J, Wang, X, Song, Q, Xu, HH, Zhang, YH. Third trimester phthalate exposure is associated with DNA methylation of growth-related genes in human placenta. Sci Rep 2016;6:33449. https://doi.org/10.1038/srep33449.Search in Google Scholar PubMed PubMed Central
44. Zhao, Y, Shi, HJ, Xie, CM, Chen, J, Laue, H, Zhang, YH. Prenatal phthalate exposure, infant growth, and global DNA methylation of human placenta. Environ Mol Mutagen 2015;56:286–92. https://doi.org/10.1002/em.21916.Search in Google Scholar PubMed
45. Xie, X, Gao, Y, Zhang, Y, Ding, Y, Shi, R, Zhou, YJ, et al.. Genome-wide analysis of DNA methylation changes in the rat ovary after prenatal exposure to di-(2-ethylhexyl)-phthalate. Zhonghua Yufang Yixue Zazhi 2012;46:840–4.Search in Google Scholar
46. Anderson, AM, Carter, KW, Anderson, D, Wise, MJ. Coexpression of nuclear receptors and histone methylation modifying genes in the testis: implications for endocrine disruptor modes of action. PLoS One 2012;7:e34158. https://doi.org/10.1371/journal.pone.0034158.Search in Google Scholar PubMed PubMed Central
47. Colon-Diaz, M, Ortiz-Santana, J, Craig, ZR. Data on the activity of DNA methyltransferase in the uteri of CD-1 mice exposed to dibutyl phthalate. Data Brief 2020;28:105061. https://doi.org/10.1016/j.dib.2019.105061.Search in Google Scholar PubMed PubMed Central
48. Li, L, Zhang, T, Qin, XS, Ge, W, Ma, HG, Sun, LL, et al.. Exposure to diethylhexyl phthalate (DEHP) results in a heritable modification of imprint genes DNA methylation in mouse oocytes. Mol Biol Rep 2014;41:1227–35. https://doi.org/10.1007/s11033-013-2967-7.Search in Google Scholar PubMed
49. Lee, J, Kim, J, Zinia, SS, Park, J, Won, S, Kim, WJ. Prenatal phthalate exposure and cord blood DNA methylation. Sci Rep 2023;13:7046. https://doi.org/10.1038/s41598-023-33002-8.Search in Google Scholar PubMed PubMed Central
50. Neier, K, Cheatham, D, Bedrosian, LD, Dolinoy, DC. Perinatal exposures to phthalates and phthalate mixtures result in sex-specific effects on body weight, organ weights and intracisternal A-particle (IAP) DNA methylation in weanling mice. J Dev Origin Health Dis 2019;10:176–87. https://doi.org/10.1017/s2040174418000430.Search in Google Scholar PubMed PubMed Central
51. Moody, L, Kougias, D, Jung, PM, Digan, I, Hong, A, Gorski, A, et al.. Perinatal phthalate and high-fat diet exposure induce sex-specific changes in adipocyte size and DNA methylation. J Nutr Biochem 2019;65:15–25. https://doi.org/10.1016/j.jnutbio.2018.11.005.Search in Google Scholar PubMed PubMed Central
52. Tindula, G, Murphy, SK, Grenier, C, Huang, Z, Huen, K, Escudero-Fung, M, et al.. DNA methylation of imprinted genes in Mexican-American newborn children with prenatal phthalate exposure. Epigenomics 2018;10:1011–26. https://doi.org/10.2217/epi-2017-0178.Search in Google Scholar PubMed PubMed Central
53. Huang, LL, Zhou, B, Ai, SH, Yang, P, Chen, YJ, Liu, C, et al.. Prenatal phthalate exposure, birth outcomes and DNA methylation of Alu and LINE-1 repetitive elements: a pilot study in China. Chemosphere 2018;206:759–65. https://doi.org/10.1016/j.chemosphere.2018.05.030.Search in Google Scholar PubMed
54. Chen, CH, Jiang, SS, Chang, IS, Wen, HJ, Sun, CW, Wang, SL. Association between fetal exposure to phthalate endocrine disruptor and genome-wide DNA methylation at birth. Environ Res 2018;162:261–70. https://doi.org/10.1016/j.envres.2018.01.009.Search in Google Scholar PubMed
55. Solomon, O, Yousefi, P, Huen, K, Gunier, RB, Escudero-Fung, M, Barcellos, LF, et al.. Prenatal phthalate exposure and altered patterns of DNA methylation in cord blood. Environ Mol Mutagen 2017;58:398–410. https://doi.org/10.1002/em.22095.Search in Google Scholar PubMed PubMed Central
56. Goodrich, JM, Dolinoy, DC, Sanchez, BN, Zhang, Z, Meeker, JD, Mercado-Garcia, A, et al.. Adolescent epigenetic profiles and environmental exposures from early life through peri-adolescence. Environ Epigenet 2016;2:dvw018. https://doi.org/10.1093/eep/dvw018.Search in Google Scholar PubMed PubMed Central
57. Yuan, B, Wu, W, Chen, M, Gu, H, Tang, Q, Guo, D, et al.. From the cover: metabolomics reveals a role of betaine in prenatal DBP exposure-induced epigenetic transgenerational failure of spermatogenesis in rats. Toxicol Sci 2017;158:356–66. https://doi.org/10.1093/toxsci/kfx092.Search in Google Scholar PubMed
58. Kamstra, JH, Sales, LB, Alestrom, P, Legler, J. Differential DNA methylation at conserved non-genic elements and evidence for transgenerational inheritance following developmental exposure to mono(2-ethylhexyl) phthalate and 5-azacytidine in zebrafish. Epigenet Chromatin 2017;10:20. https://doi.org/10.1186/s13072-017-0126-4.Search in Google Scholar PubMed PubMed Central
59. Kang, SC, Lee, BM. DNA methylation of estrogen receptor alpha gene by phthalates. J Toxicol Environ Health Part A 2005;68:1995–2003. https://doi.org/10.1080/15287390491008913.Search in Google Scholar PubMed
60. Yamagata, Y, Maekawa, R, Asada, H, Taketani, T, Tamura, I, Tamura, H, et al.. Aberrant DNA methylation status in human uterine leiomyoma. Mol Hum Reprod 2009;15:259–67. https://doi.org/10.1093/molehr/gap010.Search in Google Scholar PubMed
61. Liu, S, Yin, P, Kujawa, SA, Coon, JSt, Okeigwe, I, Bulun, SE. Progesterone receptor integrates the effects of mutated MED12 and altered DNA methylation to stimulate RANKL expression and stem cell proliferation in uterine leiomyoma. Oncogene 2019;38:2722–35. https://doi.org/10.1038/s41388-018-0612-6.Search in Google Scholar PubMed PubMed Central
62. Navarro, A, Yin, P, Monsivais, D, Lin, SM, Du, P, Wei, JJ, et al.. Genome-wide DNA methylation indicates silencing of tumor suppressor genes in uterine leiomyoma. PLoS One 2012;7:e33284. https://doi.org/10.1371/journal.pone.0033284.Search in Google Scholar PubMed PubMed Central
63. Zubrzycka, A, Zubrzycki, M, Perdas, E, Zubrzycka, M. Genetic, epigenetic, and steroidogenic modulation mechanisms in endometriosis. J Clin Med 2020;9:1309. https://doi.org/10.3390/jcm9051309.Search in Google Scholar PubMed PubMed Central
64. Yang, L, Ding, L, Ren, C, Zhang, H, Lu, J, Wang, S, et al.. A review of aberrant DNA methylation and epigenetic agents targeting DNA methyltransferase in endometriosis. Curr Drug Targets 2020;21:1047–55. https://doi.org/10.2174/1389450121666200228112344.Search in Google Scholar PubMed
65. Sato, F, Tsuchiya, S, Meltzer, SJ, Shimizu, K. MicroRNAs and epigenetics. FEBS J 2011;278:1598–609. https://doi.org/10.1111/j.1742-4658.2011.08089.x.Search in Google Scholar PubMed
66. Zota, AR, Geller, RJ, VanNoy, BN, Marfori, CQ, Tabbara, S, Hu, LY, et al.. Phthalate exposures and MicroRNA expression in uterine fibroids: the FORGE study. Epigenet Insights 2020;13:2516865720904057. https://doi.org/10.1177/2516865720904057.Search in Google Scholar PubMed PubMed Central
67. Chen, HS, Hsu, CY, Chang, YC, Chuang, HY, Long, CY, Hsieh, TH, et al.. Benzyl butyl phthalate decreases myogenic differentiation of endometrial mesenchymal stem/stromal cells through miR-137-mediated regulation of PITX2. Sci Rep 2017;7:186. https://doi.org/10.1038/s41598-017-00286-6.Search in Google Scholar PubMed PubMed Central
68. Wingfield, M, Macpherson, A, Healy, DL, Rogers, PA. Cell proliferation is increased in the endometrium of women with endometriosis. Fertil Steril 1995;64:340–6. https://doi.org/10.1016/s0015-0282(16)57733-4.Search in Google Scholar PubMed
69. Moller, B, Ronnerdag, M, Wang, G, Odlind, V, Olovsson, M. Expression of vascular endothelial growth factors and their receptors in human endometrium from women experiencing abnormal bleeding patterns after prolonged use of a levonorgestrel-releasing intrauterine system. Hum Reprod 2005;20:1410–7. https://doi.org/10.1093/humrep/deh810.Search in Google Scholar PubMed
70. Bredhult, C, Backlin, BM, Olovsson, M. Effects of some endocrine disruptors on the proliferation and viability of human endometrial endothelial cells in vitro. Reprod Toxicol 2007;23:550–9. https://doi.org/10.1016/j.reprotox.2007.03.006.Search in Google Scholar PubMed
71. An, L. Exposure to mono (2-ethylhexyl) phthalate facilitates apoptosis and pyroptosis of human endometrial microvascular endothelial cells through NLRP3 inflammasome. J Appl Toxicol 2021;41:755–64. https://doi.org/10.1002/jat.4106.Search in Google Scholar PubMed
72. Kim, YH, Kim, SH, Lee, HW, Chae, HD, Kim, CH, Kang, BM. Increased viability of endometrial cells by in vitro treatment with di-(2-ethylhexyl) phthalate. Fertil Steril 2010;94:2413–6. https://doi.org/10.1016/j.fertnstert.2010.04.027.Search in Google Scholar PubMed
73. Cho, YJ, Park, SB, Han, M. Di-(2-ethylhexyl)-phthalate induces oxidative stress in human endometrial stromal cells in vitro. Mol Cell Endocrinol 2015;407:9–17. https://doi.org/10.1016/j.mce.2015.03.003.Search in Google Scholar PubMed
74. Kim, HG, Lim, YS, Hwang, S, Kim, HY, Moon, Y, Song, YJ, et al.. Di-(2-ethylhexyl) phthalate triggers proliferation, migration, stemness, and epithelial-mesenchymal transition in human endometrial and endometriotic epithelial cells via the transforming growth factor-beta/smad signaling pathway. Int J Mol Sci 2022;23:3938. https://doi.org/10.3390/ijms23073938.Search in Google Scholar PubMed PubMed Central
75. Huang, Q, Zhang, H, Chen, YJ, Chi, YL, Dong, S. The inflammation response to DEHP through PPARgamma in endometrial cells. Int J Environ Res Public Health 2016;13:318. https://doi.org/10.3390/ijerph13030318.Search in Google Scholar PubMed PubMed Central
76. Wang, X, Shang, L, Wang, J, Wu, N, Wang, S. Effect of phthalate esters on the secretion of prostaglandins (F2alpha and E2) and oxytocin in cultured bovine ovarian and endometrial cells. Domest Anim Endocrinol 2010;39:131–6. https://doi.org/10.1016/j.domaniend.2010.03.002.Search in Google Scholar PubMed
77. Gonzalez-Martin, R, Palomar, A, Medina-Laver, Y, Quinonero, A, Dominguez, F. Endometrial cells acutely exposed to phthalates in vitro do not phenocopy endometriosis. Int J Mol Sci 2022;23:11041. https://doi.org/10.3390/ijms231911041.Search in Google Scholar PubMed PubMed Central
78. Collette, T, Maheux, R, Mailloux, J, Akoum, A. Increased expression of matrix metalloproteinase-9 in the eutopic endometrial tissue of women with endometriosis. Hum Reprod 2006;21:3059–67. https://doi.org/10.1093/humrep/del297.Search in Google Scholar PubMed
79. Chung, HW, Lee, JY, Moon, HS, Hur, SE, Park, MH, Wen, Y, et al.. Matrix metalloproteinase-2, membranous type 1 matrix metalloproteinase, and tissue inhibitor of metalloproteinase-2 expression in ectopic and eutopic endometrium. Fertil Steril 2002;78:787–95. https://doi.org/10.1016/s0015-0282(02)03322-8.Search in Google Scholar PubMed
80. Daynes, RA, Jones, DC. Emerging roles of PPARs in inflammation and immunity. Nat Rev Immunol 2002;2:748–59. https://doi.org/10.1038/nri912.Search in Google Scholar PubMed
81. Kim, SH, Cho, S, Ihm, HJ, Oh, YS, Heo, SH, Chun, S, et al.. Possible role of phthalate in the pathogenesis of endometriosis: in vitro, animal, and human data. J Clin Endocrinol Metab 2015;100:E1502–11. https://doi.org/10.1210/jc.2015-2478.Search in Google Scholar PubMed
82. Kim, Y, Kim, MR, Kim, JH, Cho, HH. Aldo-keto reductase activity after diethylhexyl phthalate exposure in eutopic and ectopic endometrial cells. Eur J Obstet Gynecol Reprod Biol 2017;215:215–19. https://doi.org/10.1016/j.ejogrb.2017.05.018.Search in Google Scholar PubMed
83. Yuan, L, Tan, L, Sun, Z, Chen, X, Li, F, He, J, et al.. Plasticizer DEHP exposure in early pregnancy affects the endometrial decidualization in mice through reducing lncRNA RP24-315D19.10 expression. Zhejiang Da Xue Xue Bao Yi Xue Ban 2023;52:1–12. https://doi.org/10.3724/zdxbyxb-2022-0669.Search in Google Scholar PubMed PubMed Central
84. Long, C, Li, Z, Liang, S, Yao, S, Zhu, S, Lu, L, et al.. Resveratrol reliefs DEHP-induced defects during human decidualization. Ecotoxicol Environ Saf 2023;258:114931. https://doi.org/10.1016/j.ecoenv.2023.114931.Search in Google Scholar PubMed
85. Huang, PC, Li, WF, Liao, PC, Sun, CW, Tsai, EM, Wang, SL. Risk for estrogen-dependent diseases in relation to phthalate exposure and polymorphisms of CYP17A1 and estrogen receptor genes. Environ Sci Pollut Res Int 2014;21:13964–73. https://doi.org/10.1007/s11356-014-3260-6.Search in Google Scholar PubMed
86. Huang, PC, Tsai, EM, Li, WF, Liao, PC, Chung, MC, Wang, YH, et al.. Association between phthalate exposure and glutathione S-transferase M1 polymorphism in adenomyosis, leiomyoma and endometriosis. Hum Reprod 2010;25:986–94. https://doi.org/10.1093/humrep/deq015.Search in Google Scholar PubMed
87. Kim, JH, Kim, SH, Oh, YS, Ihm, HJ, Chae, HD, Kim, CH, et al.. In vitro effects of phthalate esters in human myometrial and leiomyoma cells and increased urinary level of phthalate metabolite in women with uterine leiomyoma. Fertil Steril 2017;107:1061–9 e1. https://doi.org/10.1016/j.fertnstert.2017.01.015.Search in Google Scholar PubMed
88. Kim, YA, Kho, Y, Chun, KC, Koh, JW, Park, JW, Bunderson-Schelvan, M, et al.. Increased urinary phthalate levels in women with uterine leiomyoma: a case-control study. Int J Environ Res Public Health 2016;13:1247. https://doi.org/10.3390/ijerph13121247.Search in Google Scholar PubMed PubMed Central
89. Luisi, S, Latini, G, de Felice, C, Sanseverino, F, di Pasquale, D, Mazzeo, P, et al.. Low serum concentrations of di-(2-ethylhexyl)phthalate in women with uterine fibromatosis. Gynecol Endocrinol 2006;22:92–5. https://doi.org/10.1080/09513590600551312.Search in Google Scholar PubMed
90. Pollack, AZ, Buck Louis, GM, Chen, Z, Sun, L, Trabert, B, Guo, Y, et al.. Bisphenol A, benzophenone-type ultraviolet filters, and phthalates in relation to uterine leiomyoma. Environ Res 2015;137:101–7. https://doi.org/10.1016/j.envres.2014.06.028.Search in Google Scholar PubMed PubMed Central
91. Sun, J, Zhang, MR, Zhang, LQ, Zhao, D, Li, SG, Chen, B. Phthalate monoesters in association with uterine leiomyomata in Shanghai. Int J Environ Health Res 2016;26:306–16. https://doi.org/10.1080/09603123.2015.1111310.Search in Google Scholar PubMed
92. Weuve, J, Hauser, R, Calafat, AM, Missmer, SA, Wise, LA. Association of exposure to phthalates with endometriosis and uterine leiomyomata: findings from NHANES, 1999–2004. Environ Health Perspect 2010;118:825–32. https://doi.org/10.1289/ehp.0901543.Search in Google Scholar PubMed PubMed Central
93. Lee, G, Kim, S, Bastiaensen, M, Malarvannan, G, Poma, G, Caballero Casero, N, et al.. Exposure to organophosphate esters, phthalates, and alternative plasticizers in association with uterine fibroids. Environ Res 2020;189:109874. https://doi.org/10.1016/j.envres.2020.109874.Search in Google Scholar PubMed
94. Lee, J, Jeong, Y, Mok, S, Choi, K, Park, J, Moon, HB, et al.. Associations of exposure to phthalates and environmental phenols with gynecological disorders. Reprod Toxicol 2020;95:19–28. https://doi.org/10.1016/j.reprotox.2020.04.076.Search in Google Scholar PubMed
95. Fruh, V, Claus Henn, B, Weuve, J, Wesselink, AK, Orta, OR, Heeren, T, et al.. Incidence of uterine leiomyoma in relation to urinary concentrations of phthalate and phthalate alternative biomarkers: a prospective ultrasound study. Environ Int 2021;147:106218. https://doi.org/10.1016/j.envint.2020.106218.Search in Google Scholar PubMed PubMed Central
96. Pacyga, DC, Ryva, BA, Nowak, RA, Bulun, SE, Yin, P, Li, Z, et al.. Midlife urinary phthalate metabolite concentrations and prior uterine fibroid diagnosis. Int J Environ Res Public Health 2022;19:2741. https://doi.org/10.3390/ijerph19052741.Search in Google Scholar PubMed PubMed Central
97. Zhang, M, Liu, C, Yuan, XQ, Cui, FP, Miao, Y, Yao, W, et al.. Oxidatively generated DNA damage mediates the associations of exposure to phthalates with uterine fibroids and endometriosis: findings from TREE cohort. Free Radic Biol Med 2023;205:69–76. https://doi.org/10.1016/j.freeradbiomed.2023.05.029.Search in Google Scholar PubMed
98. Zota, AR, Geller, RJ, Calafat, AM, Marfori, CQ, Baccarelli, AA, Moawad, GN. Phthalates exposure and uterine fibroid burden among women undergoing surgical treatment for fibroids: a preliminary study. Fertil Steril 2019;111:112–21. https://doi.org/10.1016/j.fertnstert.2018.09.009.Search in Google Scholar PubMed PubMed Central
99. Fu, Z, Zhao, F, Chen, K, Xu, J, Li, P, Xia, D, et al.. Association between urinary phthalate metabolites and risk of breast cancer and uterine leiomyoma. Reprod Toxicol 2017;74:134–42. https://doi.org/10.1016/j.reprotox.2017.09.009.Search in Google Scholar PubMed
100. Zhu, Y, Yin, Q, Wei, D, Yang, Z, Du, Y, Ma, Y. Autophagy in male reproduction. Syst Biol Reprod Med 2019;65:265–72. https://doi.org/10.1080/19396368.2019.1606361.Search in Google Scholar PubMed
101. Kim, JH. Analysis of the in vitro effects of di-(2-ethylhexyl) phthalate exposure on human uterine leiomyoma cells. Exp Ther Med 2018;15:4972–8. https://doi.org/10.3892/etm.2018.6040.Search in Google Scholar PubMed PubMed Central
102. Iizuka, T, Yin, P, Zuberi, A, Kujawa, S, Coon, JSt, Bjorvang, RD, et al.. Mono-(2-ethyl-5-hydroxyhexyl) phthalate promotes uterine leiomyoma cell survival through tryptophan-kynurenine-AHR pathway activation. Proc Natl Acad Sci U S A 2022;119:e2208886119. https://doi.org/10.1073/pnas.2208886119.Search in Google Scholar PubMed PubMed Central
103. Kim, HJ, Kim, SH, Oh, YS, Heo, SH, Kim, KH, Kim, DY, et al.. Effects of phthalate esters on human myometrial and fibroid cells: cell culture and NOD-SCID mouse data. Reprod Sci 2021;28:479–87. https://doi.org/10.1007/s43032-020-00341-0.Search in Google Scholar PubMed
104. Murakami, K, Kotani, Y, Shiro, R, Takaya, H, Nakai, H, Matsumura, N. Endometriosis-associated ovarian cancer occurs early during follow-up of endometrial cysts. Int J Clin Oncol 2020;25:51–8. https://doi.org/10.1007/s10147-019-01536-5.Search in Google Scholar PubMed
105. Wang, Y, Nicholes, K, Shih, IM. The origin and pathogenesis of endometriosis. Annu Rev Pathol 2020;15:71–95. https://doi.org/10.1146/annurev-pathmechdis-012419-032654.Search in Google Scholar PubMed PubMed Central
106. Sharma, P, Lee, JL, Tsai, EM, Chang, Y, Suen, JL. n-Butyl benzyl phthalate exposure promotes lesion survival in a murine endometriosis model. Int J Environ Res Public Health 2021;18:3640. https://doi.org/10.3390/ijerph18073640.Search in Google Scholar PubMed PubMed Central
107. Cobellis, L, Latini, G, De Felice, C, Razzi, S, Paris, I, Ruggieri, F, et al.. High plasma concentrations of di-(2-ethylhexyl)-phthalate in women with endometriosis. Hum Reprod 2003;18:1512–5. https://doi.org/10.1093/humrep/deg254.Search in Google Scholar PubMed
108. Reddy, BS, Rozati, R, Reddy, BV, Raman, NV. Association of phthalate esters with endometriosis in Indian women. BJOG 2006;113:515–20. https://doi.org/10.1111/j.1471-0528.2006.00925.x.Search in Google Scholar PubMed
109. Reddy, BS, Rozati, R, Reddy, S, Kodampur, S, Reddy, P, Reddy, R. High plasma concentrations of polychlorinated biphenyls and phthalate esters in women with endometriosis: a prospective case control study. Fertil Steril 2006;85:775–9. https://doi.org/10.1016/j.fertnstert.2005.08.037.Search in Google Scholar PubMed
110. Itoh, H, Iwasaki, M, Hanaoka, T, Sasaki, H, Tanaka, T, Tsugane, S. Urinary phthalate monoesters and endometriosis in infertile Japanese women. Sci Total Environ 2009;408:37–42. https://doi.org/10.1016/j.scitotenv.2009.09.012.Search in Google Scholar PubMed
111. Kim, SH, Chun, S, Jang, JY, Chae, HD, Kim, CH, Kang, BM. Increased plasma levels of phthalate esters in women with advanced-stage endometriosis: a prospective case-control study. Fertil Steril 2011;95:357–9. https://doi.org/10.1016/j.fertnstert.2010.07.1059.Search in Google Scholar PubMed
112. Buck Louis, GM, Peterson, CM, Chen, Z, Croughan, M, Sundaram, R, Stanford, J, et al.. Bisphenol A and phthalates and endometriosis: the endometriosis: natural history, diagnosis and outcomes study. Fertil Steril 2013;100:162–9 e1-2. https://doi.org/10.1016/j.fertnstert.2013.03.026.Search in Google Scholar PubMed PubMed Central
113. Upson, K, Sathyanarayana, S, De Roos, AJ, Thompson, ML, Scholes, D, Dills, R, et al.. Phthalates and risk of endometriosis. Environ Res 2013;126:91–7. https://doi.org/10.1016/j.envres.2013.07.003.Search in Google Scholar PubMed PubMed Central
114. Nazir, S, Usman, Z, Imran, M, Lone, KP, Ahmad, G. Women diagnosed with endometriosis show high serum levels of diethyl hexyl phthalate. J Hum Reprod Sci 2018;11:131–6. https://doi.org/10.4103/jhrs.jhrs_137_17.Search in Google Scholar
115. Moreira Fernandez, MA, Cardeal, ZL, Carneiro, MM, Andre, LC. Study of possible association between endometriosis and phthalate and bisphenol A by biomarkers analysis. J Pharm Biomed Anal 2019;172:238–42. https://doi.org/10.1016/j.jpba.2019.04.048.Search in Google Scholar PubMed
116. Chou, YC, Chen, YC, Chen, MJ, Chang, CW, Lai, GL, Tzeng, CR. Exposure to mono-n-butyl phthalate in women with endometriosis and its association with the biological effects on human granulosa cells. Int J Mol Sci 2020;21:1794. https://doi.org/10.3390/ijms21051794.Search in Google Scholar PubMed PubMed Central
117. Yi, H, Wu, H, Zhu, W, Lin, Q, Zhao, X, Lin, R, et al.. Phthalate exposure and risk of ovarian dysfunction in endometriosis: human and animal data. Front Cell Dev Biol 2023;11:1154923. https://doi.org/10.3389/fcell.2023.1154923.Search in Google Scholar PubMed PubMed Central
118. Blount, BC, Silva, MJ, Caudill, SP, Needham, LL, Pirkle, JL, Sampson, EJ, et al.. Levels of seven urinary phthalate metabolites in a human reference population. Environ Health Perspect 2000;108:979–82. https://doi.org/10.2307/3435058.Search in Google Scholar
119. Kato, K, Silva, MJ, Reidy, JA, Hurtz, D3rd, Malek, NA, Needham, LL, et al.. Mono(2-ethyl-5-hydroxyhexyl) phthalate and mono-(2-ethyl-5-oxohexyl) phthalate as biomarkers for human exposure assessment to di-(2-ethylhexyl) phthalate. Environ Health Perspect 2004;112:327–30. https://doi.org/10.1289/ehp.6663.Search in Google Scholar PubMed PubMed Central
120. Cai, W, Yang, J, Liu, Y, Bi, Y, Wang, H. Association between phthalate metabolites and risk of endometriosis: a meta-analysis. Int J Environ Res Public Health 2019;16:3678. https://doi.org/10.3390/ijerph16193678.Search in Google Scholar PubMed PubMed Central
121. Marx, RG, Wilson, SM, Swiontkowski, MF. Updating the assignment of levels of evidence. J Bone Jt Surg Am 2015;97:1–2. https://doi.org/10.2106/jbjs.n.01112.Search in Google Scholar
122. Howick, J, Chalmers, I, Glasziou, P, Greenhalgh, T, Heneghan, C, Liberati, A. The 2011 Oxford CEBM levels of evidence (Introductory Document); 2011. Oxford Centre for Evidence-Based Medicine. Available from: http://www.cebm.net/index.aspx?o=5653.Search in Google Scholar
123. L.o.E.W.G. OCEBM. The Oxford levels of evidence; 2011, vol 2.Search in Google Scholar
124. Richardson, KA, Hannon, PR, Johnson-Walker, YJ, Myint, MS, Flaws, JA, Nowak, RA. Di (2-ethylhexyl) phthalate (DEHP) alters proliferation and uterine gland numbers in the uteri of adult exposed mice. Reprod Toxicol 2018;77:70–9. https://doi.org/10.1016/j.reprotox.2018.01.006.Search in Google Scholar PubMed
125. Kim, J, Cha, S, Lee, MY, Hwang, YJ, Yang, E, Ryou, C, et al.. Chronic low-dose nonylphenol or di-(2-ethylhexyl) phthalate has a different estrogen-like response in mouse uterus. Dev Reprod 2018;22:379–91. https://doi.org/10.12717/dr.2018.22.4.379.Search in Google Scholar
126. Somasundaram, DB, Manokaran, K, Selvanesan, BC, Bhaskaran, RS. Impact of di-(2-ethylhexyl) phthalate on the uterus of adult Wistar rats. Hum Exp Toxicol 2017;36:565–72. https://doi.org/10.1177/0960327116657601.Search in Google Scholar PubMed
127. Grande, SW, Andrade, AJ, Talsness, CE, Grote, K, Golombiewski, A, Sterner-Kock, A, et al.. 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;229:114–22. https://doi.org/10.1016/j.tox.2006.10.005.Search in Google Scholar PubMed
128. Somasundaram, DB, Selvanesan, BC, Ramachandran, I, Bhaskaran, RS. Lactational exposure to di (2-ethylhexyl) phthalate impairs the ovarian and uterine function of adult offspring rat. Reprod Sci 2016;23:549–59. https://doi.org/10.1177/1933719115607995.Search in Google Scholar PubMed
129. Kumar, N, Sharan, S, Srivastava, S, Roy, P. Assessment of estrogenic potential of diethyl phthalate in female reproductive system involving both genomic and non-genomic actions. Reprod Toxicol 2014;49:12–26. https://doi.org/10.1016/j.reprotox.2014.06.008.Search in Google Scholar PubMed
130. Kim, J, Cha, S, Lee, MY, Hwang, YJ, Yang, E, Choi, D, et al.. Chronic and low dose exposure to nonlyphenol or di(2-ethylhexyl) phthalate alters cell proliferation and the localization of steroid hormone receptors in uterine endometria in mice. Dev Reprod 2019;23:263–75. https://doi.org/10.12717/dr.2019.23.3.263.Search in Google Scholar PubMed PubMed Central
131. Shanmugam, DAS, Dhatchanamurthy, S, Leela, KA, Bhaskaran, RS. Maternal exposure to di(2-ethylhexyl) phthalate (DEHP) causes multigenerational adverse effects on the uterus of F(1) and F(2) offspring rats. Reprod Toxicol 2023;115:17–28. https://doi.org/10.1016/j.reprotox.2022.11.006.Search in Google Scholar PubMed
132. Lee, J, Chang, SH, Cho, YH, Kim, JS, Kim, H, Zaheer, J, et al.. Prenatal to peripubertal exposure to Di(2-ethylhexyl) phthalate induced endometrial atrophy and fibrosis in female mice. Ecotoxicol Environ Saf 2023;269:115798. https://doi.org/10.1016/j.ecoenv.2023.115798.Search in Google Scholar PubMed
133. Li, K, Liszka, M, Zhou, C, Brehm, E, Flaws, JA, Nowak, RA. Prenatal exposure to a phthalate mixture leads to multigenerational and transgenerational effects on uterine morphology and function in mice. Reprod Toxicol 2020;93:178–90. https://doi.org/10.1016/j.reprotox.2020.02.012.Search in Google Scholar PubMed
134. Shukla, R, Arshee, MR, Laws, MJ, Flaws, JA, Bagchi, MK, Wagoner Johnson, AJ, et al.. Chronic exposure of mice to phthalates enhances TGF beta signaling and promotes uterine fibrosis. Reprod Toxicol 2023;122:108491. https://doi.org/10.1016/j.reprotox.2023.108491.Search in Google Scholar PubMed
135. Boberg, J, Christiansen, S, Axelstad, M, Kledal, TS, Vinggaard, AM, Dalgaard, M, et al.. Reproductive and behavioral effects of diisononyl phthalate (DINP) in perinatally exposed rats. Reprod Toxicol 2011;31:200–9. https://doi.org/10.1016/j.reprotox.2010.11.001.Search in Google Scholar PubMed
136. Masutomi, N, Shibutani, M, Takagi, H, Uneyama, C, Takahashi, N, Hirose, M. Impact of dietary exposure to methoxychlor, genistein, or diisononyl phthalate during the perinatal period on the development of the rat endocrine/reproductive systems in later life. Toxicology 2003;192:149–70. https://doi.org/10.1016/s0300-483x(03)00269-5.Search in Google Scholar PubMed
Supplementary Material
The article contains supplementary material (https://doi.org/10.1515/reveh-2023-0159).
© 2024 Walter de Gruyter GmbH, Berlin/Boston
Articles in the same Issue
- Frontmatter
- Reviews
- Mercury and cadmium-induced inflammatory cytokines activation and its effect on the risk of preeclampsia: a review
- Prevalence of chronic obstructive pulmonary disease in Indian nonsmokers: a systematic review & meta-analysis
- Beyond the outdoors: indoor air quality guidelines and standards – challenges, inequalities, and the path forward
- Cadmium exposure and thyroid hormone disruption: a systematic review and meta-analysis
- New generation sequencing: molecular approaches for the detection and monitoring of bioaerosols in an indoor environment: a systematic review
- Concentration of Tetrabromobisphenol-A in fish: systematic review and meta-analysis and probabilistic health risk assessment
- The association between indoor air pollution from solid fuels and cognitive impairment: a systematic review and meta-analysis
- Phthalates and uterine disorders
- Effectiveness of educational interventions for the prevention of lead poisoning in children: a systematic review
- Association between exposure to per- and polyfluoroalkyl substances and levels of lipid profile based on human studies
- Summary of seven Swedish case reports on the microwave syndrome associated with 5G radiofrequency radiation
- Expanding the focus of the One Health concept: links between the Earth-system processes of the planetary boundaries framework and antibiotic resistance
- Exploring the link between ambient PM2.5 concentrations and respiratory diseases in the elderly: a study in the Muang district of Khon Kaen, Thailand
- Standards for levels of lead in soil and dust around the world
- Tributyltin induces apoptosis in mammalian cells in vivo: a scoping review
- The influence of geology on the quality of groundwater for domestic use: a Kenyan review
- Biological concentrations of DDT metabolites and breast cancer risk: an updated systematic review and meta-analysis
- Letter to the Editor
- Ancient medicine and famous iranian physicians
Articles in the same Issue
- Frontmatter
- Reviews
- Mercury and cadmium-induced inflammatory cytokines activation and its effect on the risk of preeclampsia: a review
- Prevalence of chronic obstructive pulmonary disease in Indian nonsmokers: a systematic review & meta-analysis
- Beyond the outdoors: indoor air quality guidelines and standards – challenges, inequalities, and the path forward
- Cadmium exposure and thyroid hormone disruption: a systematic review and meta-analysis
- New generation sequencing: molecular approaches for the detection and monitoring of bioaerosols in an indoor environment: a systematic review
- Concentration of Tetrabromobisphenol-A in fish: systematic review and meta-analysis and probabilistic health risk assessment
- The association between indoor air pollution from solid fuels and cognitive impairment: a systematic review and meta-analysis
- Phthalates and uterine disorders
- Effectiveness of educational interventions for the prevention of lead poisoning in children: a systematic review
- Association between exposure to per- and polyfluoroalkyl substances and levels of lipid profile based on human studies
- Summary of seven Swedish case reports on the microwave syndrome associated with 5G radiofrequency radiation
- Expanding the focus of the One Health concept: links between the Earth-system processes of the planetary boundaries framework and antibiotic resistance
- Exploring the link between ambient PM2.5 concentrations and respiratory diseases in the elderly: a study in the Muang district of Khon Kaen, Thailand
- Standards for levels of lead in soil and dust around the world
- Tributyltin induces apoptosis in mammalian cells in vivo: a scoping review
- The influence of geology on the quality of groundwater for domestic use: a Kenyan review
- Biological concentrations of DDT metabolites and breast cancer risk: an updated systematic review and meta-analysis
- Letter to the Editor
- Ancient medicine and famous iranian physicians