You are on page 1of 9

Reproductive Toxicology 61 (2016) 142–150

Contents lists available at ScienceDirect

Reproductive Toxicology
journal homepage: www.elsevier.com/locate/reprotox

Follicular fluid and urinary concentrations of phthalate metabolites


among infertile women and associations with in vitro fertilization
parameters
Yao-Yao Du a , Yue-Li Fang a , Yi-Xin Wang b,c , Qiang Zeng b,c , Na Guo a , Hua Zhao a ,
Yu-Feng Li a,∗
a
Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
b
Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology,
Wuhan, Hubei, PR China
c
Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental
health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China

a r t i c l e i n f o a b s t r a c t

Article history: Evidence from toxicological studies has demonstrated that phthalates can lead to reduced fertility
Received 23 September 2015 through effects on folliculogenesis, oocyte maturation and embryonic development, but human data
Received in revised form 27 March 2016 are limited. Concentrations of eight phthalate metabolites in 110 follicular fluid (FF) and urine samples
Accepted 7 April 2016
collected from 112 women attending an infertility clinic in Wuhan, China were quantified, and corre-
Available online 8 April 2016
lations between paired matrices were explored. Associations between metabolite concentrations and
in vitro fertilization (IVF) parameters were evaluated with multivariable models. Six metabolites were
Keywords:
detected in >72.73% of the FF samples. MEHP and MBP were the dominant metabolites with a median
Phthalates
Follicular fluid
level of 2.80 and 2.05 ng/mL, respectively. Significant correlations between the two matrices, urine and
IVF parameters FF, were found for MEP (rs = 0.44), and MBP (rs = 0.22). FF and urinary metabolite concentrations were
Female reproduction not associated with any IVF parameters. However, given the prevalence of phthalates exposure, further
Endocrine disruptors work is needed to elucidate the potential hazard on female reproduction.
© 2016 Elsevier Inc. All rights reserved.

1. Introduction products, or as excipients, such as coatings of medications and


dietary supplements [1–3]. Owing to the extensive use of phtha-
Phthalates are a class of man-made industrial chemicals which lates, as well as their non-covalent conjugation with the products,
have been ubiquitously applied to manufacture polyvinyl chloride the general population has been pervasively exposed to these com-
products, such as construction materials, food packaging and chil- pounds [1]. Once the phthalates enter the human body via various
dren’s toys, mainly based on their ability to impart flexibility. They routes, primarily through ingestion, they can interfere with the
are also commonly used as solvents in cosmetics and personal care endocrine systems, thus so called “endocrine-disrupting chemi-
cals (EDCs)” [1,4]. To date, detectable levels of phthalates and their
metabolites have been found in a wide range of biological fluids
including human urine, serum, semen, amniotic fluid, umbilical
Abbreviations: AFC, antral follicle count; BMI, body mass index; DEHP, cord blood and breast milk [5–8].
di(2-ethylhexyl) phthalate; EDCs, endocrine-disrupting chemicals; E2 , estradiol;
FF, follicular fluid; FSH, follicle-stimulating hormone; HCG, human chorionic
Prevalent exposure to phthalates has aroused growing pub-
gonadotropin; ICSI, intracytoplasmic sperm injection; IVF, in vitro fertilization; LOD, lic health concern based on their endocrine-disrupting potency.
the limits of detection; MBP, monobutyl phthalate; MBzP, monobenzyl phtha- The adverse effects of phthalates on male fertility have been
late; MEP, monoethyl phthalate; MEHP, mono(2-ethylhexyl) phthalate; MEHHP, well documented and extensively studied[9]. In females, on the
mono(2-ethyl-5-hydroxyhexyl) phthalate; MEOHP, mono(2-ethyl-5-oxohexyl)
other hand, the influence of phthalates on ovarian and repro-
phthalate; MMP, monomethyl phthalate; MOP, mono-n-octyl phthalate; %MEHP,
the percentage of DEHP metabolites excreted as MEHP; 2PN, two pronuclei. ductive functions remains not well understood. However, there
∗ Corresponding author at: Reproductive Medicine Center, Tongji Hospital, Tongji is accumulated evidence from experimental animal studies sug-
Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, gesting that phthalates exert reproductive toxicity by targeting
PR China. the ovary [10,11]. Exposure to phthalates has been demonstrated
E-mail addresses: yufengli64@163.com, yufengli64@tjh.tjmu.edu.cn (Y.-F. Li).

http://dx.doi.org/10.1016/j.reprotox.2016.04.005
0890-6238/© 2016 Elsevier Inc. All rights reserved.
Y.-Y. Du et al. / Reproductive Toxicology 61 (2016) 142–150 143

to disrupt folliculogenesis, steroidogenesis, oocyte maturation and 2. Materials and method


embryonic development [11–13], thus lead to reduced fertility. The
effect of di(2-ethylhexyl) phthalate (DEHP) [prototype of mono(2- 2.1. Study population and data collection
ethylhexyl) phthalate (MEHP)] on ovarian follicles and oocyte
cellular structures was evidenced by depleted primordial folli- In this prospective cohort study, 112 women seeking infertility
cle pool, increased atresia of antral follicles and observed spindle treatment at the Reproductive Medicine Center of Tongji Hospi-
abnormalities which may result in aneuploidy, miscarriages and tal, Wuhan, China, between July, 2014 and August, 2014 were
congenital defects [14,15]. A significant decrease of estradiol (E2 ) recruited. Among couples attending the infertility clinic, the female
production in a dose related pattern was observed in primary partners aged 20–45 years, diagnosed with infertility (not being
cultured granulosa cells after exposure to MEHP [16,17]. Labora- pregnant without protected intercourse for more than one year),
tory studies using in vitro maturation systems showed that MEHP and with indications for IVF or ICSI, were eligible for the study.
impeded the progression of oocyte meiosis and inhibited develop- Patients who received oocyte donation (n = 3) or used cryo-thawed
mental potential of embryos in different animal models [13,18]. oocytes (n = 2) were not included in the study. During the studying
Investigations regarding reproductive toxicity of phthalates on period, the long gonadotropin-releasing hormone (GnRH) agonist
females conducted to date have been through animal or in vitro and GnRH antagonist protocols were the main stimulation regi-
studies, whether they can execute comparable effects in humans mens used in the clinic, which accounted for nearly 90% of the
remains to be elucidated, especially, when the high dosages used treatment cycles. The clinical pregnancy rates per transfer cycle at
in laboratory experiments were considered. The hitherto published the clinic maintained at around 55%. All enrolled patients were fol-
epidemiological literature focusing on phthalates exposure and lowed up until they withdrew from the study because of achieving
female fertility is scarce. Moreover, most previous studies mea- a live birth or discontinuing the treatment due to other reasons.
sured urinary concentrations of phthalate metabolites as internal At the time of recruitment, each participant signed an informed
exposure biomarkers. Although urinary metabolites are reliable consent and completed a questionnaire collecting information
indicators of individual exposure, they may not effectively reflect about demographics, life-style, and medical and reproductive his-
the actual exposure status of the ovary. Follicular fluid (FF), the tories. Their clinical data on antral follicle count (AFC), serum peak
micro-environment that oocyte and its surrounding somatic cells E2 concentrations, and IVF outcomes, etc. were obtained from elec-
directly contact with, is critical for oocyte health [19]. Any con- tronic medical charts. AFC was determined through transvaginal
tamination with endocrine disruptors may affect developmental ultrasound on the third day of a menstrual cycle as a routine
competence of the oocyte. Therefore, FF may serve as a more suit- infertility evaluation at the clinic. The study was approved by the
able and biological relevant matrix to assess ovarian phthalate Institutional Review Board of Tongji Hospital.
exposure. Additionally, studies examining other EDCs in FF sam-
ples obtained from women undergoing IVF have demonstrated that 2.2. Ovarian stimulation
exposure to environmental chemicals can result in detectable con-
centrations in FF [20,21]. Some of these EDCs found in FF were even Patients underwent either a long GnRH agonist or a GnRH antag-
suggested to associate with adverse reproductive outcomes includ- onist protocol, according to their ovarian response. A long GnRH
ing elevated risk of fertilization and implantation failure [22,23]. agonist regimen was offered to patients with a normal ovarian
Findings from the above studies underscored the need to identify response, and pituitary suppression was achieved by daily injection
whether phthalates would present in the FF and may cause similar of triptorelin acetate (Decapeptyl; Ferring) initiating from the mid-
effects observed in experimental animal studies. luteal phase prior to the stimulation cycle. While among women
To test our hypothesis, we first aimed to quantify eight with a diminished ovarian reserve or a known poor response, a
phthalate metabolites, including monomethyl phthalate (MMP), flexible GnRH antagonist protocol was conducted by administrat-
monoethyl phthalate (MEP), mono-n-butyl phthalate (MBP), ing 0.25 mg Cetrorelix Acetate (Cetrotide; Merck-Serono) when at
MEHP, mono(2-ethyl-5-hydroxyhexyl) phthalate (MEHHP), least one follicle reached 14 mm. Recombinant follicle-stimulating
mono(2-ethyl-5-oxohexyl) phthalate (MEOHP), monobenzyl hormone (FSH) (Gonal-F, Serono or Puregon, MSD) was given to
phthalate (MBzP) and mono-n-octyl phthalate (MOP), in FF and induce ovarian stimulation starting with 150 IU/d and adjusted
urine samples from women undergoing in vitro fertilization (IVF). based on the ovarian response, as previously described [29]. Ovula-
These metabolites were chosen to determine because their parent tion was triggered by Recombinant human chorionic gonadotropin
compounds, i.e., dimethyl phthalate (DMP), diethyl phthalate (HCG) (250 mg; Ovidrel; Serono) when a mean diameter of 18 mm
(DEP), di-n-butyl phthalate (DBP), butylbenzyl phthalate (BBP), was reached by at least two leading follicles. Ovum pick-up was
di(2-ethylhexyl) phthalate (DEHP), and di-n-octyl phthalate (DOP), performed transvaginally 34–36 h after trigger with HCG.
were among the most commercially significant phthalates with
large volumes of production and widespread uses [24]. The US 2.3. IVF procedures
Environmental Protection Agency (USEPA) has also identified
the six chemicals as priority pollutants. In order to investigate Details on the IVF procedure and embryo culture have been
whether it was possible to predict concentrations of metabolites previously described [30]. Briefly, the oocytes were evaluated for
in the FF based on urinary measurements, we went further by maturity 2–4 h after ovum pick-up, as evidenced by extruding the
studying the correlations between levels in the two matrices. first polar body, and fertilized either by conventional insemina-
Finally, the associations of metabolite concentrations with IVF tion or intracytoplasmic sperm injection (ICSI). Fertilization was
parameters, comprising peak E2 levels and oocyte yielding, both checked 16–18 h after insemination, and normal fertilization was
reflecting ovarian response to hyperstimulation [25,26], as well as defined as the appearance of two pronuclei (2PN). The count of
early reproductive outcomes, such as the number of matured and 2PN relative to the amount of retrieved oocytes was referred to
fertilized oocytes, and Day 3 embryo quality, were explored. These as fertilization rate. Embryo consisted of seven to nine cells with-
early reproductive end-points which could be only observed in out multinucleation and had less than 20% fragments on day 3 after
an IVF laboratory reflect stages of reproduction that are critical to oocyte pick-up were considered as a good-quality embryo [31]. The
form a fully competent embryo and predict pregnancy outcomes percentage of optimal embryos among the total number of cleav-
[27,28]. age embryos was calculated as good-quality embryo rate. Usually,
fewer than two embryos of good quality were chosen for transfer
144 Y.-Y. Du et al. / Reproductive Toxicology 61 (2016) 142–150

on day 3, with excess embryos being cryopreserved or continuously rized with descriptive statistical analyses. Basic characteristics
cultured to the blastocyst stage. On day 5 or 6, the number of blas- were presented as mean ± SD (range) or median (IQR) or number
tocyst formation was noted and divided by the amount of embryos (%), where appropriate. Geometric means, medians and selected
cultured to blastocyst stage, referred to as blastocyst formation rate. percentiles were calculated to describe the distributions of follic-

ular fluid and urinary phthalate metabolites. A value of LOD/ 2
2.4. Sample collection and hormone measurement was assigned to samples with concentration below the LOD. To
adjust for urine dilution, concentrations of urinary metabolites
Blood serum on day 3 of the preceding menstrual cycle, together were divided by creatinine and presented as microgram per gram
with serum samples on the day of HCG administration were col- creatinine, before the analyses. Due to the relatively low detection
lected for measurement of basal FSH and peak E2 concentrations, frequencies (<50%) of MBzP in the FF specimens and MOP in both
respectively. Details of hormone measurement could be found in matrices, they were excluded from subsequent analyses. Spearman
a previous study [29]. On the day of oocyte retrieval, 110 FF and correlation analysis was conducted to explore the interrelation-
110 spot urine samples were obtained from 112 subjects. Among ships of concentrations between paired matrices. The potential
them, 108 were paired samples. All specimens were collected with associations of metabolite levels with patient and cycle character-
sterile polypropylene containers. Pooled FF from follicles >17 mm istics, including age, BMI and indicators of embryo development,
verified by ultrasound was collected. And FF contaminated with were assessed by bivariate correlation analyses.
blood or flushing medium was discarded. Following ovum pick-up, Multiple linear regression analyses were performed on both nat-
the samples were centrifuged immediately at 1500 revolution/min, ural logarithm transformed concentrations (continuous) as well
4 ◦ C for 10 min. After the treatment, the supernatant was collected as categorized data (untransformed) to evaluate the associations
and aliquoted into 2 mL specimens. Both FF and urine samples were between FF and urinary concentrations of phthalates and IVF
stored at −80 ◦ C until phthalate metabolites analysis. To adjust for parameters. Serum peak E2 levels, the number of retrieved oocytes,
urine dilution, urinary creatinine concentrations were determined mature and normally fertilized oocyte count, and the amount
with an automated clinical chemistry analyzer at the Tongji hospi- of blastocyst formed were square root transformed to normal-
tal clinical laboratory. ize their distribution, while the number of good-quality embryos
and good-quality embryo rate, with normal distribution, were
2.5. Phthalate metabolites assessment kept untransformed. For fertilization and blastocyst formation rate
which were non-normally distributed and could be expressed as a
Concentrations of eight phthalate metabolites, including MMP, proportion, namely, 2PN-zygotes among the number of retrieved
MEP, MEHHP, MBP, MEOHP, MBzP, MEHP and MOP, were quanti- oocytes and the amount of blastocyst formed relative to cultured
fied according to previously described methods [32],with minor embryo count, logistic regression was conducted as described by
modifications. Briefly, 200 ␮L of phosphoric acid was added to Rabe-Hesketh and Everit [35]. According to the distribution of
1 mL of FF specimens to terminate hydrolysis, while urine sam- the study population, metabolite concentrations (untransformed)
ples were spared this step due to lack of hydrolytic enzymes. were divided into tertiles, modeled as an ordinal categorical vari-
After being spiked with 80 ␮L of internal standards, both FF able, and assigned with an integer value of 1, 2 and 3, respectively.
and urine were incubated with 10 ␮L ␤-glucuronidase (Roche, Tests for trend were performed by entering the assigned values as a
Mannheim, Germany) at 37 ◦ C for 90 min. Following the enzy- continuous variable to explore the potential linear dose–response
matic deconjugation, the phthalate monoester compounds were relationships between tertiles of concentrations and the parame-
extracted using solid-phase extraction (SPE) cartridges (Oasis HLB, ters of interest.
Waters Co., Milford, MA). Dried extracts were then analyzed with Covariates with either biological relevance or statistical con-
high-performance liquid chromatography and tandem mass spec- siderations were selected to enter the regression models. Given
trometry (6460LC–MS, Agilent Technologies Co., Santa Clara, CA). their associations with both phthalate exposure and IVF outcomes
Apart from the study specimens, one blank and two quality con- reported in the literature [36–38], age and BMI were forced into
trols were run along with each batch of samples. The blank control regression models regardless of their effect on phthalate parame-
contained 1-mL water was used to assess the contaminations dur- ter estimate. Covariates including Day 3 serum FSH, AFC, duration
ing sample processing and analysis. Two follicular fluid samples of infertility, stimulation protocol (long GnRH agonist versus GnRH
from unknown subjects spiked with 0.5 and 5 ng/mL of the target antagonist protocol) and insemination technique (IVF versus ICSI),
phthalates, respectively, coupled with two urine samples (con- whose enter caused >10% change in effect estimates for metabo-
tained 5 and 50 ng/mL of the target phthalates), were used as quality lite regression coefficients were considered as confounders and
controls to determine the accuracy and precision of the method retained in the multivariate models. To maintain consistency,
by calculating the recovery. The linearity correlation coefficient models assessing a group of indicators representing similar out-
(R2 ) of calibration curves was >0.99, with a linear range of 0.2–10 comes, such as ovarian response or embryonic development, were
and 0.5–200 ng/mL for FF and urine samples, respectively. The lim- adjusted for the same set of covariates. To control the type I error
its of detection (LOD) of target metabolites ranged from 0.01 to rate induced by multiple comparisons, the false discovery rate
0.04 ng/mL, and the recovery for FF metabolites was 86.12-99.26%, (FDR) correction was conducted to adjust the P-values [39]. All tests
and 88.06-100.93% for urine. According to previous studies [33,34], with a P-value <0.05 were considered statistically significant. Data
the proportion of DEHP detected as the bioactive monoester in were analyzed with either the Predictive Analytics Suite Worksta-
the specimens was calculated by converting the concentrations of tion (PASW) version 22.0 (IBM Corporation, Armonk, NY) or Stata
MEHP, MEHHP and MEOHP into nanomoles per milliliter. MEHP 11 (Stata Corp LP, TX, USA).
was further divided by the sum of the three metabolites, and mul-
tiplied by 100. The computed value was expressed as %MEHP to
reflect the body burden of MEHP. 3. Results

2.6. Statistical analyses 3.1. Characteristics of study population

The demographic and clinical characteristics, cycle outcomes The patients included in the present study were on average
and the distributions of metabolite concentrations were summa- 31 ± 4.4 years old with an average BMI of 22 ± 2.5 kg/m2 , and most
Y.-Y. Du et al. / Reproductive Toxicology 61 (2016) 142–150 145

Table 1 late metabolites were highly detected in the FF (percent detectable


Demographics and clinical characteristics of the subjects (N = 112).
ranging from 72.73% to 100%), except for MBzP and MOP with a
Characteristic Data detection frequency of 43.64% and 14.55%, respectively. The high-
Age (years) 31 ± 4.4 (21–45) est amount in the FF was found for MEHP with a median level
BMI (kg/m2 ) 22 ± 2.5 (16.4–29.3) of 2.80 ng/mL, followed by MBP (median 2.05 ng/mL). The other
metabolites had much lower concentrations with median levels
Race
Han 107 (95.5) close to their respective LODs (ranging from < LOD to 0.65 ng/mL).
Other 5 (4.5) The majority of urinary metabolites were detectable in >77.27% of
Smoking
participants, with the exception of MOP (16.63%). Generally, the
Never smoked 112 (100%) urine samples had higher percentages of detectable concentra-
Ever smoked 0 tions than that in the FF. Exposure levels of urine varied widely
Gravidity by metabolite, with MBP having the highest median concentration
Yes 53 (47.3%) among the analytes (median 102.30 ␮g/g Cr) which was on aver-
Natural pregnancy 46 (86.8%) age 9-fold higher than the following MMP (median 12.51 ␮g/g Cr)
Assisted pregnancy 7 (13.2%) and MEP (median 12.17 ␮g/g Cr). In the FF samples, the concen-
No 59 (52.7%)
Duration of infertility (years) 4 (2–6)
trations of metabolites were generally two orders of magnitude
lower compared to the corresponding compounds in the urine.
IVF/ICSI treatment indication
Spearman correlation coefficients between levels of metabolites in
Tubal or pelvic factor infertility 66 (58.9)
Ovulation disorders 9 (8.0) paired FF and urine samples are shown in Table 3. Statistically sig-
Diminished ovarian reserve 7 (6.3) nificant correlations between levels in both matrices were found
Endometriosis 6 (5.4) for MEP (rs = 0.44, p < 0.01), and MBP (rs = 0.22, p < 0.05), with mod-
Uterine disorders 1 (0.9) erate to weak correlations. Nevertheless, for the other metabolites,
Male factor 22 (19.6)
no apparent correlations were observed.
Unexplained 1 (0.9)
Day 3 FSH (IU/L) 6.73 (5.64–8.16)
AFC 14 (9-19)
Peak E2 (pg/ml) 3923 (2495–5847)
3.3. Phthalate metabolite exposure and IVF parameters

Stimulation protocol
There were no associations between ln-transformed FF concen-
Long GnRH agonist protocol 95 (84.8)
GnRH-antagonist protocol 17 (15.2)
trations of phthalate metabolites in relation to the reproductive
outcomes as well as urinary results after correction for multiple
Oocyte insemination technique
comparisons (Supplementary Table S1–Table S4). The associations
IVF 80 (72.7)
ICSI 30 (27.3) of phthalate metabolite concentrations in FF with ovarian response
Oocytes retrieveda 13 (8-19) and oocyte maturation in multivariable models are presented in
Mature (MII) oocytes 11 (7-16) Table 4. Both peak serum E2 level and oocyte yield, indicators of
Maturation rate (%) 93.3 (83.8–100) ovarian response, did not correlate with any metabolites in the FF
Normal fertilized (2PN) oocytes 7 (4–11)
Fertilization rate (%) 60 (50–75)
after adjusting for age, BMI, AFC and stimulation protocol. Similarly,
Day 3 good-quality embryos 5 (2–7) the number of M II oocytes showed no associations to increasing
Good-quality embryo rate (%) 50 (33.3–63.6) metabolite concentrations. Table 5 presents the concentrations of
Blastocyst formationb 3 (1–7) FF metabolites relative to parameters of oocyte and embryo devel-
Blastocyst formation rate (%) 71.4 (50–90.9)
opment, including 2PN oocyte counts, the number of good-quality
Data are mean ± SD (range) or median (IQR) or number (%). embryos and blastocyst formed, and exhibits no evidence for any
a
Patients had oocyte retrieval procedures, n = 110. dose–response trend. No statistically significant dose-response
b
Patients had embryos cultivated to the blastocyst stage, n = 97.
patterns between phthalate metabolite concentrations and the fer-
tilization rate, the percent of good-quality embryos and the rate of
of them are Han (95.5%). None of the patients were current smok- blastocyst formation were observed after adjustment for multiple
ers or alcohol users. A total of 53 women (47.3%) had a history of testing (Table 6). To explore dose-response relationships between
pregnancy, and 86.8% got pregnant naturally. More than half of the considered parameters and the percent of DEHP excreted as the
participants underwent ART procedures because of tubal or pelvic bioactive toxicant, %MEHP were also included in the multivari-
infertility, and 19.6% due to male factor infertility. Other patient and able regression models and modeled into tertiles. However, none
cycle characteristics are presented in Table 1. Of the 112 patients, of the parameters showed any associations to %MEHP. With regard
only two women had oocyte retrieval procedures cancelled because to urinary results, there were no correlations between metabolite
of poor response to ovarian stimulation. Seven patients with less concentrations and measured reproductive outcomes in adjusted
than three good-quality cleavage embryos chose for transfer on models with correction for multiple testing (Supplementary Table
day 3 after fertilization. Six women had all day-3 embryos cry- S5–Table S7).
opreserved owing to risk of ovarian hyperstimulation syndrome
or serum progesterone elevation, thus resulting thirteen patients 4. Discussion
being excluded from the blastocyst stage analyses.
In this prospective cohort study of 112 women recruited from
3.2. Distribution of phthalate metabolite concentrations an infertility clinic, we found that the intra-ovarian environment
of the study population has been pervasively exposed to phtha-
Among the 112 participants, 108 women contributed two sam- lates despite the low concentrations. Moreover, no statistically
ples comprising one FF and one urine sample. The remaining significant associations between concentrations of FF and uri-
four participants each contributed a single sample of either kind. nary phthalate metabolites and IVF parameters, including ovarian
Thus, a total of 220 samples were subjected to metabolite concen- response and early IVF outcomes, were observed.
tration assessment. Table 2 presents the distributions of FF and Knowledge about the EDCs exposure status of the ovary is lim-
creatinine-adjusted urinary phthalate metabolites. Most of phtha- ited and of concern for reproductive risk assessment. In the current
146 Y.-Y. Du et al. / Reproductive Toxicology 61 (2016) 142–150

Table 2
Distribution of follicular fluid and urinary phthalate metabolitesa (n = 110).

Metabolites %>LOD GM Median Min Selected percentiles Max


25% 75%

MMP FF 100 0.83 0.65 0.10 0.32 1.34 133


Urine 77.27 4.31 12.51 <LOD 0.53 37.50 16444
MEP FF 96.36 0.15 0.11 0.01 0.07 0.23 11.96
Urine 100 14.80 12.17 0.12 7.26 25.79 548
MEHHP FF 100 0.18 0.15 0.02 0.09 0.04 6.68
Urine 100 10.31 9.88 1.59 6.42 14.56 286
MBP FF 88.18 1.59 2.05 <LOD 0.66 6.41 415
Urine 100 138 102.30 11.66 54.91 176 2324
MEOHP FF 72.73 0.03 0.02 <LOD 0.01 0.25 1.54
Urine 100 6.02 6.46 0.23 3.88 9.26 98.93
MBzP FF 43.64 0.03 <LOD <LOD <LOD 0.16 6.83
Urine 100 0.87 0.72 0.23 0.52 1.28 13.02
MEHP FF 93.64 1.90 2.80 <LOD 0.54 8.06 239
Urine 99.09 4.64 4.57 <LOD 2.68 9.33 113
MOP FF 14.55 0.04 <LOD <LOD <LOD <LOD 4.10
Urine 16.63 0.03 <LOD <LOD <LOD <LOD 0.61
%MEHP FF – 71.29 91.01 4.57 67.31 97.52 99.95
Urine – 21.71 23.59 1.40 16.51 30.80 68.45

%>LOD: Phthalate metabolites above the limits of detection. FF: follicular fluid. GM: geometric mean. Min: minimum. Max: maximum.
%MEHP: the percentage of DEHP metabolites excreted as MEHP.
a
Urinary phthalate metabolite concentrations were creatinine adjusted (␮g/g Cr).

Table 3
Spearman correlation coefficients of phthalate metabolites between paired follicular fluid and urine samplesa (n = 108).

Metabolites MMP MEP MEHHP MBP MEOHP MEHP

rs 0.13 0.44** 0.05 0.22* 0.12 0.11


a
Urinary phthalate metabolite concentrations were creatinine adjusted (␮g/g Cr).
**
P-value < 0.01, two-tailed.
*
P-value < 0.05, two-tailed.

study, for the first time, we determined levels of eight phthalate tices, governmental legislation and socio–economic factors, such
metabolites in 110 FF samples in a Chinese population to investi- as long-term exposure to certain phthalate-tainted food, and dif-
gate whether phthalates can reach the intra-ovarian environment ferent frequencies in cosmetics and personal care products use.
and serve as a biologically relevant marker of exposure. Our results Serving as an internal exposure biomarker, the measurement of
suggested that phthalate metabolites measured in the FF could be urinary phthalate metabolites represents exposure from all routes.
used for exposure assessment because they were detectable in the This may be the reason why the concentrations of urinary metabo-
majority of the samples, except for MBzP and MOP. We are aware lites were extraordinarily high compared to their counterparts in
of only one other publication [40] with data available on phtha- the FF. Additionally, when assessing the agreement of metabolite
late levels in FF. In contrast to our findings, MBzP and MEOHP concentrations between paired urine and FF samples, only weak
were not detected in any of the specimens. This may be attributed to moderate correlations were observed for MBP and MEP. The
to the small sample size (N = 5) of the previous study. However, reason for the lack of correlations is unclear, but it may reflect
the low concentrations found in our study were comparable to difference in the metabolic clearance of phthalates between the
theirs in which a mean level of all metabolites ranged from 1.19 to two matrices. It has been suggested that when administrated with
9.34 ng/mL. Additionally, our observation showed that MEHP and DEHP orally, the metabolites in serum reached their highest levels
MBP were the dominant metabolites in the FF, similar with their approximately 2 h after exposure and declined rapidly afterward.
findings of MEHP showing the highest amount. While concentrations in urine were still rising and achieved peak
Since access to FF is difficult which could be practically obtained levels 4 h after dosing [48]. Thus, given the fact that FF is a filtrate
through an IVF setting, we measured concentrations of phthalate of thecal capillary blood, the agreement between levels in FF and
metabolites in paired FF and urine samples to find out if levels urine samples collected at the same time may not be observed. As
in the FF could be assessed based on measurement in urine. The a result, urinary phthalate metabolite measurements may not ade-
urinary concentrations analyzed here were in the ranges of pre- quately reflect phthalate metabolite measurements in the follicular
vious studies in other regions of China with different population environment, and therefore may not be appropriate for predicting
groups [41–43], as well as a study of Japanese women [44]. Our metabolite levels in FF.
data were comparable to a recent Chinese study evaluating urinary To our knowledge, this is the first epidemiological study to
concentrations of metabolites in women with clinical pregnancy investigate the associations between FF levels of phthalate metabo-
loss [43]. The highest amount of MBP measured in our study was lites and IVF parameters. It is essential to relate our results to
in agreement with the later one. When comparing with data from experimental findings, since there is only a paucity of human data.
American and Puerto Rican women of childbearing age [45–47], Laboratory studies have established that phthalates, especially
women in the present study had similar distribution for most of the DEHP, di-n-butyl phthalate (DBP) and their respective monoesters,
metabolites, except concentrations of MBP were consistently high, adversely influence ovarian function through effect on oocyte,
while MEP exhibiting approximately 3 to 8-fold lower. Discrepan- granulosa cells and follicles at different stages, resulting in compro-
cies in the distribution of phthalates may be partially explained mised oocyte development, suppressed E2 production, accelerated
by different exposure profiles related to lifestyle, cultural prac- primordial follicle pool depletion and antral follicle atresia. How-
Y.-Y. Du et al. / Reproductive Toxicology 61 (2016) 142–150 147

Table 4
Associations between tertiles of phthalate metabolite concentrations in follicular fluid and peak E2 , number of oocytes retrieved and MII oocytes in multivariable models.

Metabolitetertilesa Peak E2 d Number of oocytes retrievedd Number of MII oocytesd

␤ (95%CI) ␤ (95%CI) ␤ (95%CI)

MMP
1b ( < 0.41) Ref Ref Ref
2 (0.41–0.92) −0.85 (−8.53, 6.83) −0.02 (−0.41, 0.36) −0.11 (−0.45, 0.24)
3 (0.93–133) −0.33 (−8.16, 7.49) 0.20 (−0.20, 0.59) 0.17 (−0.18, 0.52)
Test for trendc 0.93 0.33 0.36
MEP
1b ( < 0.088) Ref Ref Ref
2 (0.088–0.156) −5.37 (−13.37, 2.64) 0.24 (−0.16, 0.64) 0.29 (90.07, 0.65)
3 (0.157–11.96) −1.84 (−9.61, 5.94) 0.38 (−0.01, 0.77) 0.32 (−0.03, 0.67)
Test for trendc 0.66 0.06 0.08
MEHHP
1b ( < 0.11) Ref Ref Ref
2 (0.11–0.212) −2.59 (−10.25, 5.07) −0.27 (−0.65, 0.12) −0.20 (−0.55, 0.15)
3 (0.213–6.68) 0.50 (−7.28, 8.29) −0.12 (−0.51, 0.27) −0.05 (−0.40, 0.31)
Test for trendc 0.91 0.54 0.79
MBP
1b ( < 1.12) Ref Ref Ref
2 (1.12–4.13) 2.51 (−5.41, 10.42) 0.00 (−0.40, 0.41) −0.04 (−0.41, 0.32)
3 (4.14–415) −0.28 (−8.05, 7.49) −0.04 (−0.44, 0.35) 90.10 (−0.45, 0.26)
Test for trendc 0.92 0.83 0.85
MEOHP
1b ( < 0.0124) Ref Ref Ref
2 (0.0124–0.290) 6.09 (−1.50, 13.68) 0.06 (−0.36, 0.45) 0.14 (−0.22, 0.49)
3 (0.291–1.54) −1.98 (−9.67, 5.70) −0.05 (−0.45, 0.35) 0.04 (−0.32, 0.40)
Test for trendc 0.62 0.77 0.85
MEHP
1b ( < 1.08) Ref Ref Ref
2 (1.08–5.24) 3.37 (−4.54, 11.27) 0.15 (−0.25, 0.55) 0.14 (−0.22, 0.50)
3 (5.25–239) 5.52 (−2.38, 13.42) 0.22 (−0.18, 0.62) 0.21 (−0.16, 0.57)
Test for trendc 0.17 0.28 0.26
%MEHP
1b ( < 80.28) Ref Ref Ref
2 (80.28–96.35) −0.54 (−8.21, 7.12) 0.04 (−0.35, 0.43) 0.01 (−0.35, 0.37)
3 (96.36–99.95) 4.76 (−3.07, 12.59) 0.09 (−0.32, 0.49) 0.04 (−0.33, 0.40)
Test for trendc 0.23 0.67 0.84

Adjusted for age, BMI, AFC and stimulation protocol.


a
Phthalate metabolite concentrations were categorized into tertiles.
b
Reference category.
c
P-values for trend.
d
Values were square root transformed.

ever, in contrast to the animal data, no associations between FF lowing fetal and neonatal exposure in an animal model [51]. Moyer
metabolite concentrations and the considered parameters were et al. (2012) demonstrated that female rodent offspring presented
observed in the present study. Evidence from animal and in vitro early reproductive senescence arising from in utero exposure to
studies have indicated that much higher levels of phthalates were MEHP [52]. With regard to human studies, prenatal exposure to
required to induce reproductive toxicity than those detected in MEP led to decreased serum anti-Müllerian hormone (AMH) levels
our study. Using in vitro cultured human luteinized granulosa and declined incidence of polycystic ovarian morphology (PCO) in
cells, Reinsberg et al. (2009) reported that the minimal concen- adolescence, suggesting a latent effect of phthalates on the ovary
trations of MEHP required to inhibit E2 production by 50% was [53]. DEHP was shown to have an adverse impact on puberty devel-
three orders of magnitude higher than the median level we mea- opment, manifested by negative correlations between reduced
sured [17]. Similarly, exposing oocyte to 50 ␮m MEHP (identical uterus volume among adolescent girls and maternal urinary lev-
to 14 ␮g/mL after conversion) could adversely affect its nuclear els of MEHP and total DEHP [54]. Thus, exposing to phthalates
and cytoplasm maturation, contributing to reduced developmental at critical developmental stages may pose detrimental effects on
competence of oocytes, decreased cleavage rate of embryos and the female reproduction later in life and make the exposure difficult
inability to develop to the blastocyst stage [13,49]. A recent study to quantify. On the other hand, phthalates measured in FF only
demonstrated that administration of MEHP to cultured ovaries reflect a relatively short period of exposure, because the fluid does
and antral follicles at concentrations ranging from 0.2–20 ␮g/mL not generate until follicles grow into the antral phase. Therefore,
and 0.1–10 ␮g/mL, respectively, led to accelerated initial recruit- based on our observation, FF may be a feasible matrix for assess-
ment and declined levels of testosterone, estrone and estradiol [50]. ing ovarian exposure to phthalates, but it may not adequately
Therefore, the absence of associations may be partly attributed to provide informative results when studying reproductive param-
the low exposure concentrations compared to the minimum dosage eters.
reported to inducing ovarian toxicity. With regard to urinary concentrations of phthalates in rela-
Given the fact that there are a finite number of oocytes and tion to interested outcomes, a recent study by Gaskins/Hauser
oocyte development occurs from early fetal life through to adult- et al. (2015) found evidence for metabolites of DEHP in associ-
hood, the supporting microenvironment (e.g. follicular fluid) poses ation with decreased oocyte yield and amount of M II oocytes
a potential route of exposure to various EDCs [34]. It has been noted [46] in contrast to our negative findings. Their much larger
that DEHP caused increased antral follicle atresia in adulthood fol- sample size with sufficient power of test as compared to us,
148 Y.-Y. Du et al. / Reproductive Toxicology 61 (2016) 142–150

Table 5
Associations between tertiles of phthalate metabolite concentrations in follicular fluid and number of 2PN oocytes, good-quality embryos and blastocyst formation in
multivariable models.

Metabolitetertilesa Number of 2PN oocytesd Number of good-qualityembryos Number of blastocyst formationd

␤ (95%CI) ␤ (95%CI) ␤ (95%CI)

MMP
1b (< 0.41) Ref Ref Ref
2 (0.41–0.92) −0.13 (−0.47, 0.20) −0.01 (−1.34, 1.33) 0.32 (−0.15, 0.79)
3 (0.93–133) 0.25 (−0.09, 0.59) −0.22 (−1.57, 1.13) 0.42 (−0.07, 0.90)
Test for trendc 0.18 0.76 0.09

MEP
1b (< 0.088) Ref Ref Ref
2 (0.088–0.156) 0.19 (−0.17, 0.55) 0.46 (90.94, 1.86) 0.11 (−0.41, 0.62)
3 (0.157–11.96) 0.33 (−0.02, 0.67) 0.66 (−0.70, 2.02) 0.34 (−0.15, 0.84)
Test for trendc 0.06 0.34 0.16

MEHHP
1b (< 0.11) Ref Ref Ref
2 (0.11–0.212) −0.19 (−0.53, 0.15) −0.74 (−2.07, 0.59) −0.43 (−0.90, 0.05)
3 (0.213–6.68) −0.05 (−0.39, 0.30) −0.77 (−2.11, 0.58) 0.06 (−0.41, 0.54)
Test for trendc 0.79 0.26 0.80

MBP
1b (< 1.12) Ref Ref Ref
2 (1.12–4.13) −0.10 (−0.46, 0.26) −0.27 (−1.65, 1.12) −0.22 (−0.73, 0.29)
3 (4.14–415) −0.03 (−0.38, 0.32) −0.06 (−1.41, 1.29) −0.09 (−0.58, 0.40)
Test for trendc 0.87 0.95 0.76

MEOHP
1b (< 0.0124) Ref Ref Ref
2 (0.0124–0.290) 0.19 (−0.16, 0.54) 0.39 (−0.97, 1.74) 0.13 (−0.37, 0.63)
3 (0.291–1.54) 0.14 (−0.21, 0.49) −0.27 (−1.64, 1.10) −0.02 (−0.52, 0.48)
Test for trendc 0.44 0.69 0.92

MEHP
1b (< 1.08) Ref Ref Ref
2 (1.08–5.24) −0.06 (−0.41, 0.30) −0.60 (−1.98, 0.78) 0.10 (−0.41, 0.61)
3 (5.25–239) 0.09 (−0.27, 0.45) 0.07 (−1.32, 1.45) 0.16 (−0.34, 0.66)
Test for trendc 0.60 0.89 0.53

%MEHP
1b (< 80.28) Ref Ref Ref
2 (80.28–96.35) −0.06 (−0.41, 0.29) −0.03 (−1.37, 1.32) 90.16 (−0.66, 0.34)
3 (96.36k99.95) −0.10 (−0.45, 0.26) −0.42(−1.79, 0.96) −0.13 (90.63, 0.36)
Test for trendc 0.58 0.54 0.61

Adjusted for age, BMI, AFC, stimulation protocol and oocyte insemination technique (IVF/ICSI).
a
Phthalate metabolite concentrations were categorized into tertiles.
b
Reference category.
c
P-values for trend.
d
Values were square root transformed.

may partially explain the different results between the two to determine the contamination status of FF and measure the IVF
studies. Other possible reasons may rest with differences in end-points.
potential confounding (e.g., exposure to mixed chemicals), and
analytical methods (e.g., our consideration for chance finding).
5. Conclusion
Thus, additional research is needed to investigate the validity
of measuring urinary metabolites with respect to IVF out-
In conclusion, our preliminary data indicated that most phtha-
comes.
late metabolites measured in the study were highly detected in the
There are some limitations of the present study. One major limi-
ovarian FF of women undergoing IVF. However, the levels of phtha-
tation is our small sample size which may not have adequate power
lates are orders of magnitude lower than those shown to induce
to detect significant associations. Another limitation is that the
ovarian toxicity in animal studies. It has to be noted that although
study population was recruited from an infertility clinic which pre-
we did not observe any associations between phthalate metabolite
cluded extrapolation of our results to the general population. The
concentrations and IVF parameters, we cannot exclude their pos-
single spot urine and single measurement of FF used for exposure
sible influences on female reproduction which could be detected
assessment may cause exposure misclassifications owing to their
from a larger sample analysis. Thus, further study with a larger
relatively short biological half-life. However, it has been reported
cohort would be warranted to verify the findings of the present
that when modeled into categories based on exposure levels, a sin-
study.
gle measure of spot urine is possible to represent the exposure
extent over several months [55,56]. Finally, exposure to mixtures
of environmental pollutants with diverse toxicity may influence Conflict of interest
the interpretation of our data. Despite the aforementioned limi-
tations, this study is strengthened by our prospective study design The authors declare that there are no competing financial inter-
and the application of an IVF setting which provided us an approach ests.
Y.-Y. Du et al. / Reproductive Toxicology 61 (2016) 142–150 149

Table 6
Associations between tertiles of phthalate metabolite concentrations in follicular fluid and fertilization rate, good-quality embryos rate and blastocyst formation rate in
multivariable models.

Metabolitetertilesa Fertilization rated Good-quality embryo ratee Blastocyst formation ratef

OR (95%CI) ␤ (95%CI) OR (95%CI)

MMP
1b (< 0.41) Ref Ref Ref
2 (0.41–0.92) 0.86 (0.67, 1.11) 0.01 (−0.09, 0.12) 1.49 (0.99, 2.24)
3 (0.93–133) 1.16 (0.89, 1.49) −0.03 (−0.13, 0.08) 1.58 (1.03, 2.44)
Test for trendc 0.46 0.66 0.14

MEP
1b (< 0.088) Ref Ref Ref
2 (0.088–0.156) 1.11 (0.85, 1.45) 0.07 (−0.04, 0.18) 1.00 (0.66, 1.53)
3 (0.157–11.96) 1.25 (0.95, 1.63) 0.03 (−0.08, 0.13) 1.83 (1.18, 2.85)
Test for trendc 0.35 0.66 0.07

MEHHP
1b (< 0.11) Ref Ref Ref
2 (0.11–0.212) 1.06 (0.82, 1.38) −0.01 (−0.12, 0.10) 1.20 (0.91, 1.59)
3 (0.213–6.68) 1.11 (0.86, 1.43) −0.11 (−0.22, 0.00) 1.29 (0.97, 1.70)
Test for trendc 0.50 0.35 0.63

MBP
1b (< 1.12) Ref Ref Ref
2 (1.12–4.13) 0.77 (0.60, 1.00) 0.06 (−0.05, 0.17) 0.81 (0.53, 1.22)
3 (4.14–415) 0.98 (0.76, 1.27) −0.04 (−0.15, 0.07) 0.90 (0.60, 1.37)
Test for trendc 0.95 0.63 0.63

MEOHP
1b (< 0.0124) Ref Ref Ref
2 (0.0124–0.290) 1.07 (0.82, 1.39) 0.01 (−0.10, 0.11) 1.18 (0.78, 1.79)
3 (0.291–1.54) 1.20(0.93, 1.55) −0.06 (−0.17, 0.05) 1.23 (0.81, 1.86)
Test for trendc 0.35 0.63 0.46

MEHP
1b (< 1.08) Ref Ref Ref
2 (1.08–5.24) 0.80 (0.61, 1.05) −0.06 (−0.17, 0.05) 0.89 (0.57, 1.40)
3 (5.25–239) 0.86 (0.66, 1.13) −0.09 (−0.20, 0.02) 0.79 (0.52, 1.22)
Test for trendc 0.50 0.39 0.46

%MEHP
1b (< 80.28) Ref Ref Ref
2 (80.28–96.35) 0.78 (0.60, 1.01) 0.02 (−0.09, 0.13) 0.90 (0.58, 1.40)
3 (96.36–99.95) 0.71 (0.55, 0.92) −0.05 (−0.16, 0.06) 0.76 (0.50, 1.16)
Test for trendc 0.07 0.63 0.44
a
Phthalate metabolite concentrations were categorized into tertiles.
b
Reference category.
c
FDR adjusted P-values for trend.
d
Adjusted for age and BMI.
e
Adjusted for age, BMI, AFC, duration of infertility and oocyte insemination technique (IVF/ICSI).
f
Adjusted for age, BMI, FSH and duration of infertility.

Transparency document References

The Transparency document associated with this article can be [1] U. Heudorf, V. Mersch-Sundermann, Angerer J. Phthalates, toxicology and
exposure, Int. J. Hyg. Environ. Health 210 (2007) 623–634.
found in the online version. [2] T. Schettler, Human exposure to phthalates via consumer products, Int. J.
Androl. 29 (2006) 134–139, discussion 81-5.
[3] K.E. Kelley, S. Hernandez-Diaz, E.L. Chaplin, R. Hauser, A.A. Mitchell,
Identification of phthalates in medications and dietary supplement
Acknowledgements formulations in the United States and Canada, Environ. Health Perspect. 120
(2012) 379–384.
We sincerely thank the nurses and technicians in the Reproduc- [4] Z.R. Craig, W. Wang, J.A. Flaws, Endocrine-disrupting chemicals in ovarian
function: effects on steroidogenesis, metabolism and nuclear receptor
tive Medicine Center of Tongji Hospital in Wuhan for clinical data signaling, Reproduction 142 (2011) 633–646.
and sample collection. This study was supported by the National [5] J. Hogberg, A. Hanberg, M. Berglund, S. Skerfving, M. Remberger, A.M. Calafat,
Natural Science Foundation of China (grant 81571508), the Natural et al., Phthalate diesters and their metabolites in human breast milk, blood or
serum, and urine as biomarkers of exposure in vulnerable populations,
Science Foundation of Hubei Province (grant 2014CFA069) and the
Environ. Health Perspect. 116 (2008) 334–339.
Self-dependent Innovation Research Funding of Huazhong Univer- [6] M.J. Silva, J.A. Reidy, A.R. Herbert, J.L. Preau Jr., L.L. Needham, A.M. Calafat,
sity of Science and Technology (grant 2014KXYQ017). Detection of phthalate metabolites in human amniotic fluid, Bull. Environ.
Contam. Toxicol. 72 (2004) 1226–1231.
[7] Y. Huang, J. Li, J.M. Garcia, H. Lin, Y. Wang, P. Yan, et al., Phthalate levels in
cord blood are associated with preterm delivery and fetal growth parameters
Appendix A. Supplementary data in Chinese women, PLoS One 9 (2014) e87430.
[8] K. Kato, M.J. Silva, L.L. Needham, A.M. Calafat, Quantifying phthalate
metabolites in human meconium and semen using automated off-line
Supplementary data associated with this article can be found, solid-phase extraction coupled with on-line SPE and isotope-dilution
in the online version, at http://dx.doi.org/10.1016/j.reprotox.2016. high-performance liquid chromatography–tandem mass spectrometry, Anal.
04.005. Chem. 78 (2006) 6651–6655.
150 Y.-Y. Du et al. / Reproductive Toxicology 61 (2016) 142–150

[9] V.R. Kay, M.S. Bloom, W.G. Foster, Reproductive and developmental effects of [34] R. Hauser, Urinary phthalate metabolites and semen quality: a review of a
phthalate diesters in males, Crit. Rev. Toxicol. 44 (2014) 467–498. potential biomarker of susceptibility, Int. J. Androl. 31 (2008) 112–117.
[10] T. Lovekamp-Swan, B.J. Davis, Mechanisms of phthalate ester toxicity in the [35] S.E.B. Rabe-Hesketh, A Handbook of Statistical Analyses Using Stata, Fourth
female reproductive system, Environ. Health Perspect. 111 (2003) 139–145. ed., Chapman and Hall/CRC, 2016.
[11] P.R. Hannon, J.A. Flaws, The effects of phthalates on the ovary, Front. [36] S. Bhattacharya, A. Maheshwari, J. Mollison, Factors associated with failed
Endocrinol. (Lausanne) 6 (2015) 8. treatment: an analysis of 121,744 women embarking on their first IVF cycles,
[12] M.K. Anas, C. Suzuki, K. Yoshioka, S. Iwamura, Effect of mono-(2-ethylhexyl) PLoS One 8 (2013) e82249.
phthalate on bovine oocyte maturation in vitro, Reprod. Toxicol. 17 (2003) [37] E.E. Hatch, J.W. Nelson, M.M. Qureshi, J. Weinberg, L.L. Moore, M. Singer, et al.,
305–310. Association of urinary phthalate metabolite concentrations with body mass
[13] D. Grossman, D. Kalo, M. Gendelman, Z. Roth, Effect of di-(2-ethylhexyl) index and waist circumference: a cross-sectional study of NHANES data,
phthalate and mono-(2-ethylhexyl) phthalate on in vitro developmental 1999–2002, Environ. Health 7 (2008) 27.
competence of bovine oocytes, Cell Biol. Toxicol. 28 (2012) 383–396. [38] V. Rittenberg, S. Seshadri, S.K. Sunkara, S. Sobaleva, E. Oteng-Ntim, T.
[14] P.R. Hannon, K.E. Brannick, W. Wang, R.K. Gupta, J.A. Flaws, Di(2-ethylhexyl) El-Toukhy, Effect of body mass index on IVF treatment outcome: an updated
phthalate inhibits antral follicle growth, induces atresia, and inhibits steroid systematic review and meta-analysis, Reprod. Biomed. Online 23 (2011)
hormone production in cultured mouse antral follicles, Toxicol. Appl. 421–439.
Pharmacol. 284 (2015) 42–53. [39] N. Pike, Using false discovery rates for multiple comparisons in ecology and
[15] X.F. Zhang, L.J. Zhang, L. Li, Y.N. Feng, B. Chen, J.M. Ma, et al., Diethylhexyl evolution, Methods Ecol. Evol. 2 (2011) 278–282.
phthalate exposure impairs follicular development and affects oocyte [40] S.P. Krotz, S.A. Carson, C. Tomey, J.E. Buster, Phthalates and bisphenol do not
maturation in the mouse, Environ. Mol. Mutagen. 54 (2013) 354–361. accumulate in human follicular fluid, J. Assist. Reprod. Genet. 29 (2012)
[16] T.N. Lovekamp, B.J. Davis, Mono-(2-ethylhexyl) phthalate suppresses 773–777.
aromatase transcript levels and estradiol production in cultured rat granulosa [41] Y. Guo, Q. Wu, K. Kannan, Phthalate metabolites in urine from China, and
cells, Toxicol. Appl. Pharmacol. 172 (2001) 217–224. implications for human exposures, Environ. Int. 37 (2011) 893–898.
[17] J. Reinsberg, P. Wegener-Toper, K. van der Ven, H. van der Ven, D. [42] C.Y. Chen, Y.Y. Chou, Y.M. Wu, C.C. Lin, S.J. Lin, C.C. Lee, Phthalates may
Klingmueller, Effect of mono-(2-ethylhexyl) phthalate on steroid production promote female puberty by increasing kisspeptin activity, Hum. Reprod. 28
of human granulosa cells, Toxicol. Appl. Pharmacol. 239 (2009) 116–123. (2013) 2765–2773.
[18] A. Dalman, H. Eimani, H. Sepehri, S.K. Ashtiani, M.R. Valojerdi, P. [43] D. Mu, F. Gao, Z. Fan, H. Shen, H. Peng, J. Hu, Levels of phthalate metabolites in
Eftekhari-Yazdi, et al., Effect of mono-(2-ethylhexyl) phthalate (MEHP) on urine of pregnant women and risk of clinical pregnancy loss, Environ. Sci.
resumption of meiosis, in vitro maturation and embryo development of Technol. 49 (2015) 10651–10657.
immature mouse oocytes, Biofactors 33 (2008) 149–155. [44] Y. Suzuki, M. Niwa, J. Yoshinaga, Y. Mizumoto, S. Serizawa, H. Shiraishi,
[19] D.A. Dumesic, D.R. Meldrum, M.G. Katz-Jaffe, R.L. Krisher, W.B. Schoolcraft, Prenatal exposure to phthalate esters and PAHs and birth outcomes, Environ.
Oocyte environment: follicular fluid and cumulus cells are critical for oocyte Int. 36 (2010) 699–704.
health, Fertil. Steril. 103 (2015) 303–316. [45] Y. Guo, J. Weck, R. Sundaram, A.E. Goldstone, G.B. Louis, K. Kannan, Urinary
[20] Y. Ikezuki, O. Tsutsumi, Y. Takai, Y. Kamei, Y. Taketani, Determination of concentrations of phthalates in couples planning pregnancy and its
bisphenol A concentrations in human biological fluids reveals significant association with 8-hydroxy-2’-deoxyguanosine, a biomarker of oxidative
early prenatal exposure, Hum. Reprod. 17 (2002) 2839–2841. stress: longitudinal investigation of fertility and the environment study,
[21] J.D. Meeker, S.A. Missmer, L. Altshul, A.F. Vitonis, L. Ryan, D.W. Cramer, et al., Environ. Sci. Technol. 48 (2014) 9804–9811.
Serum and follicular fluid organochlorine concentrations among women [46] R. Hauser, A.J. Gaskins, I. Souter, K.W. Smith, L.E. Dodge, S. Ehrlich, et al.,
undergoing assisted reproduction technologies, Environ. Health 8 (2009) 32. Urinary phthalate metabolite concentrations and reproductive outcomes
[22] E.M. Petro, J.L. Leroy, A. Covaci, E. Fransen, D. De Neubourg, A.C. Dirtu, et al., among women undergoing fertilization: results from the EARTH study,
Endocrine-disrupting chemicals in human follicular fluid impair in vitro Environ. Health Perspect. (2015).
oocyte developmental competence, Hum. Reprod. 27 (2012) 1025–1033. [47] D.E. Cantonwine, J.F. Cordero, L.O. Rivera-Gonzalez, L.V. Anzalota Del Toro,
[23] P.I. Johnson, L. Altshul, D.W. Cramer, S.A. Missmer, R. Hauser, J.D. Meeker, K.K. Ferguson, B. Mukherjee, et al., Urinary phthalate metabolite
Serum and follicular fluid concentrations of polybrominated diphenyl ethers concentrations among pregnant women in Northern Puerto Rico: distribution,
and in-vitro fertilization outcome, Environ. Int. 45 (2012) 9–14. temporal variability, and predictors, Environ. Int. 62 (2014) 1–11.
[24] A. Gomez-Hens, M.P. Aguilar-Caballos, Social and economic interest in the [48] H.M. Koch, H.M. Bolt, R. Preuss, J. Angerer, New metabolites of
control of phthalic acid esters, Trac-Trends in Anal. Chemi. 22 (2003) 847–857. di(2-ethylhexyl)phthalate (DEHP) in human urine and serum after single oral
[25] R. Orvieto, E. Zohav, S. Scharf, J. Rabinson, S. Meltcer, E.Y. Anteby, et al., The doses of deuterium-labelled DEHP, Arch. Toxicol. 79 (2005) 367–376.
influence of estradiol/follicle and estradiol/oocyte ratios on the outcome of [49] D. Kalo, Z. Roth, Effects of mono(2-ethylhexyl)phthalate on cytoplasmic
controlled ovarian stimulation for in vitro fertilization, Gynecol. Endocrinol. maturation of oocytes - The bovine model, Reprod. Toxicol. 53 (2015)
23 (2007) 72–75. 141–151.
[26] A. Demirol, T. Gurgan, Comparison of microdose flare-up and antagonist [50] P.R. Hannon, K.E. Brannick, W. Wang, J.A. Flaws, Mono(2-ethylhexyl)
multiple-dose protocols for poor-responder patients: a randomized study, phthalate accelerates early folliculogenesis and inhibits steroidogenesis in
Fertil. Steril. 92 (2009) 481–485. cultured mouse whole ovaries and antral follicles, Biol. Reprod. 92 (2015) 120.
[27] G.M. Jones, A.O. Trounson, N. Lolatgis, C. Wood, Factors affecting the success [51] S.W. Grande, A.J. Andrade, C.E. Talsness, K. Grote, A. Golombiewski, A.
of human blastocyst development and pregnancy following in vitro Sterner-Kock, et al., A dose-response study following in utero and lactational
fertilization and embryo transfer, Fertil. Steril. 70 (1998) 1022–1029. exposure to di-(2-ethylhexyl) phthalate (DEHP): reproductive effects on adult
[28] S.K. Sunkara, V. Rittenberg, N. Raine-Fenning, S. Bhattacharya, J. Zamora, A. female offspring rats, Toxicology 229 (2007) 114–122.
Coomarasamy, Association between the number of eggs and live birth in IVF [52] B. Moyer, M.L. Hixon, Reproductive effects in F1 adult females exposed in
treatment: an analysis of 400 135 treatment cycles, Hum. Reprod. 26 (2011) utero to moderate to high doses of mono-2-ethylhexylphthalate (MEHP),
1768–1774. Reprod. Toxicol. 34 (2012) 43–50.
[29] B. Xu, Z. Li, H. Zhang, L. Jin, Y. Li, J. Ai, et al., Serum progesterone level effects [53] R. Hart, D.A. Doherty, H. Frederiksen, J.A. Keelan, M. Hickey, D. Sloboda, et al.,
on the outcome of in vitro fertilization in patients with different ovarian The influence of antenatal exposure to phthalates on subsequent female
response: an analysis of more than 10,000 cycles, Fertil Steril. 97 (2012) reproductive development in adolescence: a pilot study, Reproduction 147
1321–1327, e1-4. (2014) 379–390.
[30] L. Zhu, Q. Xi, H. Zhang, Y. Li, J. Ai, L. Jin, Blastocyst culture and [54] P.H. Su, C.K. Chang, C.Y. Lin, H.Y. Chen, P.C. Liao, C.A. Hsiung, et al., Prenatal
cryopreservation to optimize clinical outcomes of warming cycles, Reprod. exposure to phthalate ester and pubertal development in a birth cohort in
Biomed. Online 27 (2013) 154–160. central Taiwan: a 12-year follow-up study, Environ. Res. 136 (2015) 324–330.
[31] N. Nasiri, P. Eftekhari-Yazdi, An overview of the available methods for [55] R. Hauser, J.D. Meeker, S. Park, M.J. Silva, A.M. Calafat, Temporal variability of
morphological scoring of pre-implantation embryos in in vitro fertilization, urinary phthalate metabolite levels in men of reproductive age, Environ.
Cell J. 16 (2015) 392–405. Health Perspect. 112 (2004) 1734–1740.
[32] L. You, Y.X. Wang, Q. Zeng, M. Li, Y.H. Huang, Y. Hu, et al., Semen phthalate [56] M. Fisher, T.E. Arbuckle, R. Mallick, A. LeBlanc, R. Hauser, M. Feeley, et al.,
metabolites, spermatozoa apoptosis, and DNA damage: a cross-sectional Bisphenol A and phthalate metabolite urinary concentrations: daily and
study in china, Environ. Sci. Technol. 49 (2015) 3805–3812. across pregnancy variability, J. Expo. Sci. Environ. Epidemiol. 25 (2015)
[33] R. Hauser, J.D. Meeker, N.P. Singh, M.J. Silva, L. Ryan, S. Duty, et al., DNA 231–239.
damage in human sperm is related to urinary levels of phthalate monoester
and oxidative metabolites, Hum. Reprod. 22 (2007) 688–695.

You might also like