• 沒有找到結果。

Association between perfluoroalkyl substances and reproductive hormones in adolescents and young adults

N/A
N/A
Protected

Academic year: 2021

Share "Association between perfluoroalkyl substances and reproductive hormones in adolescents and young adults"

Copied!
42
0
0

加載中.... (立即查看全文)

全文

(1)

Association between perfluoroalkyl substances and reproductive hormones in adolescents and young adults

Meng-Shan Tsaia, Chien-Yu Linb,c, Ching-Chun Lina, Mei-Huei Chend, Sandy H.J.

Hsue, Kuo-Liong Chienf,g, Fung-Chang Sungh, Pau-Chung Chena,i,j,*, Ta-Chen Sug,j,*

a Institute of Occupational Medicine and Industrial Hygiene, College of Public Health,

National Taiwan University, Taipei 100, Taiwan

b Department of Internal Medicine, En Chu Kong Hospital, New Taipei City 237,

Taiwan

c School of Medicine, Fu Jen Catholic University, Taipei County 242, Taiwan

d Department of Pediatrics, National Taiwan University Hospital Yun-Lin Branch

e Department of Laboratory Medicine, National Taiwan University Hospital, Taipei,

Taiwan

f Institute of Epidemiology and Preventive Medicine, College of Public Health,

National Taiwan University, Taipei 100, Taiwan

g Department of Internal Medicine, National Taiwan University Hospital, Taipei 100,

Taiwan

h Department of Public Health, College of Public Health, China Medical University,

Taichung 404, Taiwan 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 1

(2)

i Department of Public Health, National Taiwan University College of Public Health,

Taipei, Taiwan

j Department of Environmental and Occupational Medicine, National Taiwan

University Hospital, Taipei 100, Taiwan

*Corresponding author: Ta-Chen Su, MD, PhD

Department of Internal Medicine, National Taiwan University Hospital, 7 Chung-Shan South Road, Taipei 10055, Taiwan.

Tel: +886-2-23123456 ext 66719 Fax: +886-2-23712361 Email: tachensu@ntu.edu.tw

**Co-correspondence:

Pau-Chung Chen, MD, PhD

Institute of Occupational Medicine and Industrial Hygiene, National Taiwan University College of Public Health, #17 Syujhou Road, Taipei 10055, Taiwan.

Telephone: +886-2-3366-8088 Fax: +886-2-3366-8734 Email: pchen@ntu.edu.tw 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38

(3)

Running title: Perfluoroalkyl substances and reproductive hormones. Acknowledgements

This study was supported by grants from National Health Research Institute of

Taiwan (EX97-9721PC, EX97-9821PC, X97-9921PC, EX95-9531PI, EX95-9631PI

and EX95-9731PI), from Ministry of Science and Technology

(101-2314-B-002-184-MY3) and (99-2314-B-385-001-MY3 and 102-2314-B-002-166-(101-2314-B-002-184-MY3), and from

Taiwan and the Environmental Medicine Collaboration Center (NTUH 103 A123 and

104 A123). This work was supported in part by the 3rd core facility at National

Taiwan University Hospital.

Conflict of Interest

We declare there is no conflict of interest regarding this manuscript, including financial, consultant, institutional, and other relationships that might lead to bias or a conflict of interest. 39 40 41 42 43 44 45 46 47 48 49 50 51 5

(4)

Highlights

Serum concentration of PFAS was associated with reproductive hormone based on a

young population.

The concentrations of PFOA, PFOS, and PFUA associated with reproductive

hormone significantly.

Reproductive hormones of females, ages 12-17, were significantly influenced by

PFAS concentrations. 52 53 54 55 56 57 58

(5)

Abstract

Background: Few studies have explored the association between perfluoroalkyl

substances (PFAS) and reproductive hormones in adolescents and young adults.

Objectives: This study aimed to investigate the association of PFAS with

reproductive hormones in adolescents and young adults.

Methods: We recruited 540 subjects aged 12-30 years from a 1992-2000 mass urine

screening population and established a cohort from 2006 to 2008 via invitations by

mail or/and telephone. Serum PFAS levels were analyzed with a Waters ACQUITY

UPLC system coupled with a Waters Quattro Premier XE triple quadrupole mass

spectrometer. Serum reproductive hormone levels were measured by

immunoluminometric assay with an Architect random access assay system. PFAS

levels were divided into different percentiles according to their detection limits in the

multiple regression models to analyze associations between reproductive hormone

levels and exposure with PFAS.

Results: The adjusted mean serum level of sex hormone-binding globulin (SHBG)

decreased significantly in association with t he <50th, 50–75, 75-90 and >90th

percentile categories of perfluorooctanoic acid (PFOA) compared with a reference

category for the females in the 12-17-year-old group. The follicle-stimulating

hormone (FSH) levels were significantly decreased in association with the different

1 59 60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 9

(6)

percentile categories of perfluorooctane sulfonate (PFOS) in the male 12-17-year-old

group and the different percentile categories of perfluoroundecanoic acid (PFUA) in

the female 12-17-year-old group. The serum FSH levels in the females aged 12-17

were also decreased in association with the different percentile categories of PFUA.

On the other hand, there was a significantly negative association between the different

percentile categories of PFOS and the serum testosterone level among the female

12-17-year-old group.

Conclusions: We found that the serum concentrations of PFOA, PFOS, and PFUA

were negatively associated with the serum levels of SHBG, FSH, and testosterone in

the young Taiwanese population and that these effects were the strongest in the

females aged 12-17.Further studies are needed to determine whether these

associations are causal.

Key words: perfluoroalkyl substances, reproductive hormone, adolescent, young

adults 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92

(7)

Introduction

Perfluoroalkyl substances (PFAS), such as perfluorooctanoic acid (PFOA) and

perfluorooctane sulfonate (PFOS), are highly bio-accumulative environmental

pollutants (Austin et al. 2003) that, which are widely used for both industrial and

everyday purposes (Lau et al. 2004). Although food (including migration from

packaging and cookware) and drinking water are the primary sources of PFAS

exposure in humans, additional exposure routes include air and dust (Haug et al.

2011).Therefore, these substances are widespread in the environment, affecting

wildlife and humans (Kannan et al. 2004). In addition, they have been associated with

adverse health effects. Until now, the majority of animal studies have associated

exposure to PFAS with developmental deficits (Lau et al. 2004), neurotoxicity

(Johansson et al. 2008), and immunotoxicity (Keil et al. 2008). Epidemiological

research has shown an association among PFAS exposure, child development (Chen

et al. 2013), higher thyroid levels (Lin et al. 2013b), and immune system function

(Dong et al. 2013).

PFOS and PFOA are well-recognized endocrine disruptors that have antagonistic

effects on the synthesis of steroid hormone (Zhao et al. 2010). Reproductive

hormones are important for the reproductive system because they play pivotal roles in

both male and female puberty development and are crucial to growth and the

3 93 94 95 96 97 98 99 100 101 102 103 104 105 106 107 108 109 110 111 13

(8)

functioning of a broad range of tissues (Kjeldsen and Bonefeld-Jorgensen 2013). In

general, the average age of the onset of puberty is ten years for girls and twelve years

for boys, suggesting that the female onset of puberty occurs earlier than that of males

(Sorensen et al. 2012). Few prior epidemiological studies have investigated the impact

of PFAS on the human reproductive system, and results have been inconsistent. J

Joensen et al. selected 247 healthy young Danish men with a median age of 19 years

to examine the association between reproductive hormones, semen quality and PFAS

in the general population (Joensen et al. 2013). They found that the serum PFOS

concentration was negatively associated with testosterone levels. Additionally,

Lopez-Espinosa et al. have demonstrated that the delay of puberty in children is correlated

with PFOS and PFOA levels based on the level of testosterone or oestradiol and the

self-reported status (Lopez-Espinosa et al. 2011). A nested case-control study reported

that PFAS exposure during pregnancy was not associated with age at menarche in a

British cohort (Christensen et al. 2011). Furthermore, a recent study of a pregnant

Danish cohort has found that higher levels of PFOA exposure in utero may be

associated with a later age of menarche compared with lower levels of exposure

(Kristensen et al. 2013). In animals, PFOA has been associated with decreased serum

testosterone levels in Leydig cell adenomas (Cook et al. 1992) and increased

oestradiol levels in rodents (Biegel et al. 1995). The mechanism of the endocrine-112 113 114 115 116 117 118 119 120 121 122 123 124 125 126 127 128 129 130

(9)

disrupting activities of PFAS has been discussed by Lau et al., who have reported that

this estrogenic effect is mediated via the oestrogen receptor pathway (Lau et al. 2007).

Studies investigating the impact of PFAS on human reproductive health are limited

and controversial and have mainly focused on PFOA and PFOS. Furthermore, there

are few studies investigating the health effects of PFAS in adolescents. Therefore, the

aim of this study was to assess the association between PFAS and reproductive

hormones in young adolescents and young adults.

5 131 132 133 134 135 136 137 17

(10)

Materials and methods

Subjects and data collection

The study groups were established from a 1992-2000 mass urine screening

population of individuals attending grades 1-12 in Taiwan (Wei et al. 2003). From

2006 to 2008, we invited students in the Taipei area to participate in the study and a

follow-up health examination at the National Taiwan University Hospital. Trained

assistants and nurses invited these subjects by mail or/and telephone to undergo the

health examination and complete a questionnaire.The information has been detailed

in previous studies (Lin et al. 2013a; Lin et al. 2013b; Su et al. 2014). Out of 7,097

subjects living in Taipei, 790 students completed the follow-up health examination,

including the collection of a blood sample and the completion of a questionnaire, but

we did not measure the serum PFAS or reproductive hormone levels of all of the

subjects due to the collection of limited serum samples. Among the 790 students, 145

had an insufficient serum sample volume for PFAS measurement, and an additional

105 had an insufficient reproductive hormone measurement. Thus, 540 subjects with

both PFAS and reproductive hormone measurements were included in the final

analysis. This study was approved by the Ethics Committee of the National Taiwan

University Hospital (Research Ethics Committee, NTUH). All of the participants and

their parents (for the child and adolescent participants) signed informed consent 138 139 140 141 142 143 144 145 146 147 148 149 150 151 152 153 154 155 156

(11)

documents upon enrolment in the study.

A questionnaire, including patient age, gender, BMI, lifestyle (drinking, eating, and

exercise), household income, and dietary intake (high fat and high sugar), were

recorded during the follow-up examinations performed from 2006 to 2008. The

subjects were separated by gender and were subdivided by age into 12-17-year-old

and 18-30-year-old groups according to the clinical (Moore et al. 2013) definition of

adolescence as the period from 12 to 17 years of age. Moreover, the general

population has been reported to experience puberty between the ages of 10 and 17

years (Parent et al. 2003). Smoking status (active smoker, passive smoker or has never

smoked) and alcohol intake (current alcohol consumption or no alcohol consumption)

were determined via the questionnaire and categorized. Household income groups

were categorized as above 50,000 New Taiwan dollars (NTD) (equivalent to $1,600

USD) per month or below. Weight and height were measured during the follow-up

health examination. Body mass index (BMI) was determined as the weight (in

kilograms) divided by the square of the height (in metres). Exercise was assessed

based on the presence or absence of current exercise habits. A high-sugar diet was

defined as one in which subjects consumed sweet foods and soft drinks at a frequency

of four times a week or more. Subjects that consumed ham, fatty foods, and fast foods

at a frequency of four times a week or more were categorized as having a high-fat

7 157 158 159 160 161 162 163 164 165 166 167 168 169 170 171 172 173 174 175 21

(12)

diet.

Exposure assessment

All of the plasma samples were stored at -80 ℃ prior to PFAS analysis. A previous

study has reported a fast and sensitive ultra-high performance liquid

chromatography/tandem mass spectrometry method to determine PFAS levels (Lien

et al. 2011). We first analysed the levels of 12 PFAS, including potassium

perfluorohexanesulfonate (PFHxS), perfluoroheptanoic acid (PFHpA),

perfluorononanoic acid (PFNA), perfluorooctanoic acid (PFOA), perfluorooctyl

sulfonate (PFOS), perfluorodecanoic acid (PFDeA), perfluoroundecanoic acid

(PFUA), perfluorododecanoic acid (PFDoA), 2-(N-methyl-perfluorooctane

sulfonamido) acetic acid (Me–PFOSA–AcOH), 2-(N-ethylperfluorooctane

sulfonamido) acetic acid (Et–PFOSA–AcOH), perfluorohexanoic acid (PFHxA), and

perfluorooctane sulfonamide (PFOSA). However, the levels of eight of these PFAS

were more than 70% below the limit of quantitation (LOQ). Therefore, only PFOA,

PFOS, PFNA, and PFUA were used for the final analysis. The details of the analytical

methods have been previously described (Lien et al. 2011; Lin et al. 2013b). The

samples were first vortexed for homogeneity for 30 seconds. An additional 30-second

vortex was performed following the addition of 100 µL of 1% formic acid (pH=2.8)

to the 100-µl plasma samples. Eighty microliters of methanol and 20 µl of a 0.375 176 177 178 179 180 181 182 183 184 185 186 187 188 189 190 191 192 193 194

(13)

ng/mL internal standard (13C8-PFOA) in methanol were mixed with the solution, and

the mixture was then sonicated for 20 minutes and centrifuged at 14,000 rpm for 20

minutes. Before analysis, the supernatant was collected and filtered through a 0.22 µm

PVDF syringe filter. One hundred microliters of bovine plasma with standard

calibration solutions were prepared as described above. The concentrations of the

specific analytes were equivalent to 0.05 mL in 300 ng/mL bovine plasma with a

fixed amount of internal standard (75 ng/mL). All samples were analysed using a

Waters ACQUITY UPLC System (Waters Corporation, Milford, MA) coupled with a

Waters Quattro Premier XE triple quadrupole mass spectrometer (Waters

Corporation, Milford, MA). The limit of quantitation (LOQ) for PFOA and PFUA

was 1.5 ng/mL, that for PFOS was 0.22 ng/mL, and that for PFNA was 0.75 ng/mL.

No PFOS and trace background amounts of PFOA (up to 1.5 ng/mL), PFNA (up to

0.75 ng/mL), and PFUA (up to 3 ng/mL) were detected in the blank samples.

Therefore, the reported PFOA, PFNA, and PFUA concentrations were corrected by

subtracting the background levels of the blank. The PFAS concentration was below

the detection limits (37.4% for PFOA, 1.5% for PFOS, 55% for PFNA and 25% for

PFUA); therefore, we used a proxy value of half of the detection limit.

Assessment of reproductive hormones

The samples were stored at -80 ℃ after centrifugation until analysis was performed.

9 195 196 197 198 199 200 201 202 203 204 205 206 207 208 209 210 211 212 213 25

(14)

The reproductive hormones and PFAS were analysed at the same time. The serum

concentrations of reproductive hormones, including sex hormone-binding globulin

(SHBG), oestrogen (E2), follicle-stimulating hormone (FSH), luteinizing hormone

(LH), testosterone (T), and free testosterone (free T), were measured by

immunoluminometric assay with an Architect random access assay system (Abbott

Diagnostics, Abbott Park, IL). The intra-assay coefficients of variation of these

measurements were all below 10%, and the inter-assay coefficients of variation were

all below 15%.

Statistical analysis

The PFAS concentrations were described as the geometric mean and geometric

standard deviation. The relationships among the PFAS were assessed using the

Spearman correlation coefficient. The Mann–Whitney U test or Kruskal–Wallis test

was used to evaluation the relationships among the PFAS variables and categorical

variables. We divided each PFAS concentration into different categories for linear

regression analysis due to the high percentage of PFAS below the detection limit

(37.4% for PFOA, 1.5% for PFOS, 55% for PFNA and 25% for PFUA). The PFOA

levels were divided into the 50th (the reference category), 75th, and 90th percentiles,

and the PFOS and PFUA levels were divided into the 25th (the reference category),

50th, and 75th percentiles. Additionally, the PFNA levels were divided into the 60th 214 215 216 217 218 219 220 221 222 223 224 225 226 227 228 229 230 231 232

(15)

(the reference category) and 90th percentiles. The SHBG, E2, FSH, LH, T, and free T

levels were skewed and were thus log-transformed in the regression models.

Covariates, including age, sex, BMI, and high-fat diet intake, were considered to be

significant predictive outcomes in the regression model because they changed the

estimates by >10%. Other covariates, such as household income, alcohol

consumption, smoking status, high-sugar diet, and exercise, were considered but were

not included in the final model.

E Each PFAS model was analysed separately. SAS (version 9.3; SAS Institute Inc.,

Cary, NC, USA) was used to perform all statistical analyses and a p-value of <0.05

was considered to be statistically significant.

11 233 234 235 236 237 238 239 240 241 242 29

(16)

Results

The basic characteristics of the sample population are shown in Table 1. The

geometric mean and geometric standard deviation of the concentrations of PFOA,

PFOS, PFNA, and PFUA were 2.74 (2.95) ng/mL, 7.78 (2.40) ng/mL, 1.10 (3.55)

ng/mL, and 5.84 (2.88) ng/mL, respectively. Among the 540 subjects, 330 were

female and 210 were male. The males had a significantly higher mean concentration

of PFOS than the females (p<0.001). The 18- to 30-year-old groups had higher mean

serum concentrations of PFOS than the 12- to 17-year-old groups (p<0.05). The

subjects with a higher BMI (≥24) also had a higher PFOS concentration than those

with a BMI of below 24 (P<0.05). In addition, the PFOS and PFNA serum

concentrations were higher in those consuming a high-fat diet (p<0.05). The

reproductive hormone levels are shown in Supplementary Data (Table 2).

The association between the serum level of PFAS and that of sex hormone-binding

globulin (SHBG) is shown in Table 3. The serum level of SHBG in the females

decreased significantly in association with the percentile categories (<50th, 50–75,

75-90 and >90th percentiles) of PFOA (p for trend <0.05). The association between

FSH and the serum level of PFAS after adjustments for covariates is listed in Table 4.

The mean serum follicle-stimulating hormone (FSH) level was significantly decreased

(p for trend <0.05) in association with the different percentile categories of PFOS in 243 244 245 246 247 248 249 250 251 252 253 254 255 256 257 258 259 260 261

(17)

the male subjects aged 12-17. The female serum FSH level in the 12-17-year-old

group was also decreased (p for trend <0.01) across the different percentile categories

of PFUA. As shown in Table 5, there was a significantly negative association (p for

trend <0.05) among the percentile categories of PFOS with the female serum

testosterone level in the 12-17-year-old group, but the other three PFAS were not

associated with testosterone. However, the serum levels of PFOA, PFOS, PFNA, and

PFUA were not associated with the oestrogen level. The concentration of PFAS was

also not associated with the LH level. Moreover, the serum level of PFAS was not

associated with free testosterone. The association between PFAS and the reproductive

hormones of the subpopulations are shown in Tables S1-S7. There were no

associations between PFAS and the reproductive hormone levels stratified by BMI

and high-fat diet.

13 262 263 264 265 266 267 268 269 270 271 272 273 33

(18)

Discussion

In this study, we found that the serum level of PFAS was associated with

reproductive hormones in the younger age groups, especially PFOA, PFOS, and

PFUA. Moreover, the reproductive hormones of the females were significantly

influenced by the PFOS and PFUA concentrations. However, we did not find that an

elevation in the PFAS level was associated with the oestrogen or LH level. To our

knowledge, this is the first study to assess the serum levels of PFAS and reproductive

hormones in a cohort of adolescent and young adults; however, the effects were

determined to be minute and subclinical.

Our results revealed that the serum PFOA, PFOS, and PFUA concentrations were

higher in the 12-17-year-old group compared with the 18-30-year-old group. Due to

the existence of different growth stages, we divided the subjects into two groups and

examined the differences between them. The subjects in the 12-17-year-old group

were within the age range of puberty, which is a complex and immature stage driven

by the endocrine system (Den Hond and Schoeters 2006). One review study has

investigated the effects of endocrine disrupters on puberty and has found that

exposure to endocrine disrupters may be associated with puberty disturbances,

including delayed male and accelerated female puberty (Maranghi and Mantovani

2012). Another study has also revealed that in utero expose to high levels of PFOA 274 275 276 277 278 279 280 281 282 283 284 285 286 287 288 289 290 291 292

(19)

may delay menarche in females (Kristensen et al. 2013). PFAS act as an endocrine

disrupter; therefore, the 12-17-year-old group was more vulnerable than the

18-30-year-old group to this exposure, which resulted in obvious effects. One study has also

found that the age of puberty onset is correlated with PFOS and PFOA concentrations

(Lopez-Espinosa et al. 2011). However, the primary mechanism is unclear and

requires further investigation.

We found that the PFOA, PFOS and PFUA concentrations were negatively

associated with reproductive hormones, except LH, oestrogen, and free testosterone.

Although there are few human studies investigating associations between PFAS

exposure and reproductive hormones in adolescent and young adults, Joensen et al.

have reported negative associations between PFOS exposure and testosterone in

young men (median age of 19 years) (Joensen et al. 2013). A study of puberty

evaluating males of a similar age found a relationship between reduced odds of the

onset of puberty with an increase in the serum level of PFOS in boys, using total

testosterone as a puberty indicator (Lopez-Espinosa et al. 2011). However, additional

studies have found no association between PFOS and reproductive hormones, except

for SHBG in the spouses of pregnant women (Specht et al. 2012), and a relationship

has also been detected between PFOS and PFOA and testosterone levels in males

(Joensen et al. 2013; Raymer et al. 2012). Thus, associations between PFAS exposure

15 293 294 295 296 297 298 299 300 301 302 303 304 305 306 307 308 309 310 311 37

(20)

and reproductive hormones have been reported; however, the possible effects of

PFAS exposure are inconsistent. The observations of the current study may provide

novel information in this regard.

Most relevant published studies have focused primarily on male adult populations

and have rarely examined the effects of PFAS on female study populations (Joensen

et al. 2013; Lopez-Espinosa et al. 2011; Raymer et al. 2012). Our study revealed that

female reproductive hormones were affected by PFAS. Female SHBG decreased with

increasing serum PFOA, FSH decreased with increasing serum PFUA, and

testosterone decreased with increasing PFOS. One possible explanation for these

findings is that females usually experience the onset of puberty earlier than males and

are thus more vulnerable to the effects of PFAS than males. An animal study reported

that the levels of ammonium perfluorooctanoate (C8), administered by gavage for 14

days to adult male rats, were associated with increased oestradiol levels (Biegel et al.

1995). However, we did not observe similar results. We found that male FSH

decreased with increasing serum PFOS. The human reproductive system is

complicated and is influenced by the hypothalamus and pituitary glands. FSH is

primarily regulated by the hypothalamus and pituitary glands and responds to

development, whereas LH serves as a secondary trigger of this regulation (Moore et

al. 2013). Therefore, we observed a significant association between FSH and PFUA 312 313 314 315 316 317 318 319 320 321 322 323 324 325 326 327 328 329 330

(21)

and PFOS ; however, we did not observe an association between reproductive

hormones and PFAS. However, more studies, including not only animal studies but

also epidemiological studies of humans, are needed to assess the association between

PFAS and reproductive hormones.

We also found that SHBG in the female subjects was inversely associated with

PFOA. Recently, a review has investigated the bioaccumulation of PFAS and has

concluded that PFAS has a tendency towards protein binding; however, there are

some mechanisms that still need to be explained (Ng and Hungerbuhler 2014). On the

other hand, hormone levels and mechanisms such as enzyme activity (Zhao et al.

2010), cell membrane fluidity and permeability (Hu et al. 2003), and tissue

distribution (Ng and Hungerbuhler 2014) are quite complex; moreover, the

physiology of hormone regulation is rather complicated in adolescents, and most

underlying mechanisms are not well understood. However, the females in our

population did not exhibit variations in their hormone levels due to menstruation,

which is the most important limitation that may have influenced our results. These

results suggest that PFAS have endocrine-disrupting properties. Additional studies are

needed to examine the related health outcomes.

Subgroup analysis did not indicate that the hormone levels varied due to a higher

BMI or high-fat diet intake across the different PFAS concentrations; however, high

17 331 332 333 334 335 336 337 338 339 340 341 342 343 344 345 346 347 348 349 41

(22)

BMI and high-fat diet intake are risk factors for hormone imbalance. High doses of

PFOS and PFOA have been associated with weight loss in animal studies (Hines et al.

2009; Thibodeaux et al. 2003). A human epidemiological study has shown that PFOS,

PFOA, and PFNA have few meaningful associations with body weight (Nelson et al.

2010), but other studies have explored prenatal PFOA exposure and body weight later

in life (Halldorsson et al. 2012). An additional study has shown that obesity causes

alterations in the reproductive hormones of male adolescents (Zhang et al. 2013). An

animal study has revealed that high-fat diets induce the early onset of reproductive

function in female rats (Fungfuang et al. 2013). Important contributors to PFOA

intake are derived from different types of dietary routes (Noorlander et al. 2011).

Therefore, more studies are needed to investigate the associations between body

weight and reproductive hormones and between dietary intake and reproductive

hormones across different PFAS concentrations.

The strengths of our study included the use of a complete set of PFAS data and the

measurement of a variety of reproductive hormones in adolescents and young adults.

The measurements of PFAS have been validated (Lin et al. 2013b) and have been

demonstrated to have different health effects in this cohort (Lin et al. 2011; Lin et al.

2013a; Lin et al. 2013b; Su et al. 2014). This study also had several limitations. The

main limitation was that we did not consider menarche status or examine any other 350 351 352 353 354 355 356 357 358 359 360 361 362 363 364 365 366 367 368

(23)

puberty indicators, although they may influence hormone levels and results.

Additionally, although reproductive hormone levels in women vary according to

menarche status, we considered these levels to be random sampling results. Second,

other environmental pollutants that may impact both PFAS and reproductive hormone

levels were not measured in this study. Third, causality cannot be confirmed because

this is a cross-sectional study. Furthermore, we did not take into account medications

that may impact reproductive hormones; however, more than 95% of the participants

self-reported no significant clinical diseases and no medication history. Finally, our

study subjects were obtained from a mass urine screening population in Taipei. We

cannot extrapolate the same association to the general population; however, this fact

does not necessarily negate the clinical relevance of this study.

In conclusion, we found that PFAS levels were associated with reproductive

hormone levels, particularly the higher PFOA and lower SHBG levels, higher PFUA

and PFOS and lower FSH levels, and higher PFOS and lower testosterone levels in

the adolescents and young adults. Although the observed potential biological effects

on humans were low and subclinical in the study population, the effects observed

among the females and the 12-17-year-old group are of particular interest. Further

long-term cohort studies are needed to clarify whether these associations are causal.

19 369 370 371 372 373 374 375 376 377 378 379 380 381 382 383 384 385 386 45

(24)

References

Austin ME, Kasturi BS, Barber M, Kannan K, MohanKumar PS, MohanKumar SM.

2003. Neuroendocrine effects of perfluorooctane sulfonate in rats. Environmental

health perspectives 111:1485-1489.

Biegel LB, Liu RC, Hurtt ME, Cook JC. 1995. Effects of ammonium

perfluorooctanoate on leydig cell function: In vitro, in vivo, and ex vivo studies.

Toxicology and applied pharmacology 134:18-25.

Chen MH, Ha EH, Liao HF, Jeng SF, Su YN, Wen TW, et al. 2013. Perfluorinated

compound levels in cord blood and neurodevelopment at 2 years of age.

Epidemiology 24:800-808.

Christensen KY, Maisonet M, Rubin C, Holmes A, Calafat AM, Kato K, et al. 2011.

Exposure to polyfluoroalkyl chemicals during pregnancy is not associated with

offspring age at menarche in a contemporary british cohort. Environment

international 37:129-135.

Cook JC, Murray SM, Frame SR, Hurtt ME. 1992. Induction of leydig cell adenomas

by ammonium perfluorooctanoate: A possible endocrine-related mechanism.

Toxicology and applied pharmacology 113:209-217.

Den Hond E, Schoeters G. 2006. Endocrine disrupters and human puberty.

International journal of andrology 29:264-271; discussion 286-290. 387 388 389 390 391 392 393 394 395 396 397 398 399 400 401 402 403 404 405

(25)

polyfluoroalkyl concentrations, asthma outcomes, and immunological markers in

a case-control study of taiwanese children. Environmental health perspectives

121:507-513, 513e501-508.

Fungfuang W, Nakao N, Nakada T, Yokosuka M, Saito TR. 2013. Early onset of

reproductive function in female rats treated with a high-fat diet. The Journal of

veterinary medical science / the Japanese Society of Veterinary Science

75:523-526.

Halldorsson TI, Rytter D, Haug LS, Bech BH, Danielsen I, Becher G, et al. 2012.

Prenatal exposure to perfluorooctanoate and risk of overweight at 20 years of

age: A prospective cohort study. Environmental health perspectives

120:668-673.

Haug LS, Huber S, Schlabach M, Becher G, Thomsen C. 2011. Investigation on per-

and polyfluorinated compounds in paired samples of house dust and indoor air

from norwegian homes. Environmental science & technology 45:7991-7998.

Hines EP, White SS, Stanko JP, Gibbs-Flournoy EA, Lau C, Fenton SE. 2009.

Phenotypic dichotomy following developmental exposure to perfluorooctanoic

acid (pfoa) in female cd-1 mice: Low doses induce elevated serum leptin and

insulin, and overweight in mid-life. Molecular and cellular endocrinology

304:97-105.

Hu W, Jones PD, DeCoen W, King L, Fraker P, Newsted J, et al. 2003. Alterations in

21 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 49

(26)

cell membrane properties caused by perfluorinated compounds. Comparative

biochemistry and physiology Toxicology & pharmacology : CBP 135:77-88.

Joensen UN, Veyrand B, Antignac JP, Blomberg Jensen M, Petersen JH, Marchand P,

et al. 2013. Pfos (perfluorooctanesulfonate) in serum is negatively associated

with testosterone levels, but not with semen quality, in healthy men. Human

reproduction 28:599-608.

Johansson N, Fredriksson A, Eriksson P. 2008. Neonatal exposure to perfluorooctane

sulfonate (pfos) and perfluorooctanoic acid (pfoa) causes neurobehavioural

defects in adult mice. Neurotoxicology 29:160-169.

Kannan K, Corsolini S, Falandysz J, Fillmann G, Kumar KS, Loganathan BG, et al.

2004. Perfluorooctanesulfonate and related fluorochemicals in human blood

from several countries. Environmental science & technology 38:4489-4495.

Keil DE, Mehlmann T, Butterworth L, Peden-Adams MM. 2008. Gestational

exposure to perfluorooctane sulfonate suppresses immune function in b6c3f1

mice. Toxicological sciences : an official journal of the Society of Toxicology

103:77-85.

Kjeldsen LS, Bonefeld-Jorgensen EC. 2013. Perfluorinated compounds affect the

function of sex hormone receptors. Environmental science and pollution research

international 20:8031-8044. 427 428 429 430 431 432 433 434 435 436 437 438 439 440 441 442 443 444 445

(27)

2013. Long-term effects of prenatal exposure to perfluoroalkyl substances on

female reproduction. Human reproduction 28:3337-3348.

Lau C, Butenhoff JL, Rogers JM. 2004. The developmental toxicity of perfluoroalkyl

acids and their derivatives. Toxicology and applied pharmacology 198:231-241.

Lau C, Anitole K, Hodes C, Lai D, Pfahles-Hutchens A, Seed J. 2007. Perfluoroalkyl

acids: A review of monitoring and toxicological findings. Toxicological sciences

: an official journal of the Society of Toxicology 99:366-394.

Lien GW, Wen TW, Hsieh WS, Wu KY, Chen CY, Chen PC. 2011. Analysis of

perfluorinated chemicals in umbilical cord blood by ultra-high performance

liquid chromatography/tandem mass spectrometry. Journal of chromatography B,

Analytical technologies in the biomedical and life sciences 879:641-646.

Lin CY, Wen LL, Lin LY, Wen TW, Lien GW, Chen CY, et al. 2011. Associations

between levels of serum perfluorinated chemicals and adiponectin in a young

hypertension cohort in taiwan. Environmental science & technology

45:10691-10698.

Lin CY, Lin LY, Wen TW, Lien GW, Chien KL, Hsu SH, et al. 2013a. Association

between levels of serum perfluorooctane sulfate and carotid artery intima-media

thickness in adolescents and young adults. International journal of cardiology

168:3309-3316.

Lin CY, Wen LL, Lin LY, Wen TW, Lien GW, Hsu SH, et al. 2013b. The

23 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 53

(28)

associations between serum perfluorinated chemicals and thyroid function in

adolescents and young adults. Journal of hazardous materials 244-245:637-644.

Lopez-Espinosa MJ, Fletcher T, Armstrong B, Genser B, Dhatariya K, Mondal D, et

al. 2011. Association of perfluorooctanoic acid (pfoa) and perfluorooctane

sulfonate (pfos) with age of puberty among children living near a chemical plant.

Environmental science & technology 45:8160-8166.

Maranghi F, Mantovani A. 2012. Targeted toxicological testing to investigate the role

of endocrine disrupters in puberty disorders. Reproductive toxicology

33:290-296.

Moore KL, Persaud TVN, Torchia MG. 2013. The developing human : Clinically

oriented embryology. 9th ed. Philadelphia, PA:Saunders/Elsevier.

Nelson JW, Hatch EE, Webster TF. 2010. Exposure to polyfluoroalkyl chemicals and

cholesterol, body weight, and insulin resistance in the general u.S. Population.

Environmental health perspectives 118:197-202.

Ng CA, Hungerbuhler K. 2014. Bioaccumulation of perfluorinated alkyl acids:

Observations and models. Environmental science & technology 48:4637-4648.

Noorlander CW, van Leeuwen SP, Te Biesebeek JD, Mengelers MJ, Zeilmaker MJ.

2011. Levels of perfluorinated compounds in food and dietary intake of pfos and

pfoa in the netherlands. Journal of agricultural and food chemistry 59:7496-7505. 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484 485

(29)

The timing of normal puberty and the age limits of sexual precocity: Variations

around the world, secular trends, and changes after migration. Endocrine reviews

24:668-693.

Raymer JH, Michael LC, Studabaker WB, Olsen GW, Sloan CS, Wilcosky T, et al.

2012. Concentrations of perfluorooctane sulfonate (pfos) and perfluorooctanoate

(pfoa) and their associations with human semen quality measurements.

Reproductive toxicology 33:419-427.

Sorensen K, Mouritsen A, Aksglaede L, Hagen CP, Mogensen SS, Juul A. 2012.

Recent secular trends in pubertal timing: Implications for evaluation and

diagnosis of precocious puberty. Hormone research in paediatrics 77:137-145.

Specht IO, Hougaard KS, Spano M, Bizzaro D, Manicardi GC, Lindh CH, et al. 2012.

Sperm DNA integrity in relation to exposure to environmental perfluoroalkyl

substances - a study of spouses of pregnant women in three geographical regions.

Reproductive toxicology 33:577-583.

Su TC, Liao CC, Chien KL, Hsu SH, Sung FC. 2014. An overweight or obese status

in childhood predicts subclinical atherosclerosis and

prehypertension/hypertension in young adults. Journal of atherosclerosis and

thrombosis. J. Atheroscler. Thromb. 21 (11), 1170–1182.

Thibodeaux JR, Hanson RG, Rogers JM, Grey BE, Barbee BD, Richards JH, et al.

2003. Exposure to perfluorooctane sulfonate during pregnancy in rat and mouse.

25 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 57

(30)

I: Maternal and prenatal evaluations. Toxicological sciences : an official journal

of the Society of Toxicology 74:369-381.

Wei JN, Sung FC, Lin CC, Lin RS, Chiang CC, Chuang LM. 2003. National

surveillance for type 2 diabetes mellitus in taiwanese children. JAMA : the

journal of the American Medical Association 290:1345-1350.

Zhang LD, Li HC, Gao M, Wang L, Deng Q, Shi T, et al. 2013. [sexual development

characteristics and sex hormone levels in obese male adolescents]. Zhonghua nan

ke xue = National journal of andrology 19:434-438.

Zhao B, Hu GX, Chu Y, Jin X, Gong S, Akingbemi BT, et al. 2010. Inhibition of

human and rat 3beta-hydroxysteroid dehydrogenase and 17beta-hydroxysteroid

dehydrogenase 3 activities by perfluoroalkylated substances. Chemico-biological

interactions 188:38-43.

Table 1.

Basic demographics of the sample subjects including geometric mean (geometric SD) of PFAS concentrations.

No. PFOA (ng/ml) PFOS (ng/ml) PFNA (ng/ml) PFUA (ng/ml)

Total 540 2.74 (2.95) 7.78 (2.40) 1.10 (3.55) 5.84 (2.88) Sex Male 210 2.75 (2.93) 8.97 (2.59)** 1.21 (3.86) 5.89 (2.95) Female 330 2.73 (2.97) 7.11 (2.25)** 1.03 (3.36) 5.81 (2.83) Age (years) 12-17 95 3.03 (2.98) 7.12 (1.95)* 0.98 (3.53) 6.42 (3.06) 18-30 445 2.68 (2.94) 7.93 (2.49)* 1.12 (3.56) 5.72 (2.84) 507 508 509 510 511 512 513 514 515 516 517 518 519 520

(31)

<50000NTD per month 220 2.58 (2.99) 7.43 (2.40) 1.01 (3.50) 5.89 (2.88) ≥50000NTD per month 319 2.84 (2.92) 8.04 (2.41) 1.16 (3.58) 5.78 (2.88) Smoking status Never smoked 452 2.78 (2.97) 7.62 (2.41) 1.04 (3.47) 5.76 (2.85) Passive smoker 13 2.12 (2.89) 9.94 (1.66) 1.46 (4.70) 7.02 (3.21) Active smoker 70 2.65 (2.83) 8.43 (2.51) 1.38 (3.85) 6.18 (3.01)

Current alcohol consumption

No 465 2.76 (2.97) 7.65 (2.40) 1.04 (3.51) 5.78 (2.90)

Yes 67 2.69 (2.86) 8.72 (2.52) 1.40 (3.68) 6.27 (2.72)

Body mass index (kg/m2)

<24 417 2.88 (2.90) 7.47 (2.38)* 1.08 (3.53) 5.76 (2.84)

≥24 123 2.31 (3.09) 8.97 (2.44)* 1.16 (3.64) 6.12 (3.02)

Exercise

No 336 2.74 (2.89) 7.23 (2.49) 1.14 (3.50) 5.72 (2.90)

Yes 144 2.68 (2.88) 8.20 (2.22) 1.23 (3.82) 5.75 (2.95)

High sugar diet

No 342 2.58(2.92) 7.54(2.24) 1.15(3.52) 5.78(2.92)

Yes 138 3.10(2.76) 7.44(2.85) 1.19(3.79) 5.60(2.92)

High fat diet

No 443 2.66 (2.88) 7.36 (2.38)* 1.11 (3.48)* 5.63 (2.90)

Yes 37 3.48 (2.87) 9.60 (2.70)* 1.94 (4.67)* 7.03 (3.01)

PFAS, perfluoroalkyl substances; PFOA, perfluorooctanoic acid; PFOS, perfluorooctane sulfonate; PFNA, perfluorononanoic acid; PFUA, perfluoroundecanoic acid; NTD, new Taiwan dollars.

* p value < 0.05 **p value <0.01

27

(32)

Table 2.

Distribution of reproductive hormones in the whole study group.

Mean SD Median Percentile 05 Percentile 95

All E2 (pg/mL) 85.3 84.2 48.2 48.2 290.5 FSH (mIU/mL) 4.70 2.67 4.07 4.07 8.71 LH (mIU/mL) 3.82 4.61 2.48 2.48 11.6 SHBG (nmol/L) 42.7 26.0 36.8 36.8 91.7 Testosterone (ng/dL) 253.9 286.7 54.4 54.4 789 Free Testosterone (ng/dL) 5.40 6.43 0.86 0.86 17.0 Male 12-17 E2 (pg/mL) 36.1 10.0 36.0 36.0 53.3 FSH (mIU/mL) 4.15 1.96 4.02 4.02 8.13 LH (mIU/mL) 1.60 0.87 1.41 1.41 3.54 SHBG (nmol/L) 28.6 14.1 29.1 29.1 50.6 Testosterone (ng/dL) 508.2 181.8 537.5 537.5 820.0 Free Testosterone (ng/dL) 10.7 3.74 10.4 10.4 17.4 Female 12-17 E2 (pg/mL) 89.6 81.8 55.7 55.7 257 FSH (mIU/mL) 5.41 2.05 5.47 5.47 8.63 LH (mIU/mL) 4.58 3.75 3.70 3.70 12.9 SHBG (nmol/L) 45.0 21.2 43.6 43.6 81.8 Testosterone (ng/dL) 42.7 20.0 38.4 38.4 87.8 Free Testosterone (ng/dL) 0.66 0.34 0.59 0.59 1.28

(33)

Male 18-30 E2 (pg/mL) 36.9 10.9 35.2 35.2 54.7 FSH (mIU/mL) 3.72 1.91 3.25 3.25 7.53 LH (mIU/mL) 1.75 0.74 1.57 1.57 3.23 SHBG (nmol/L) 27.3 11.9 25.9 25.9 50.9 Testosterone (ng/dL) 599.8 167.2 590.5 590.5 890.5 Free Testosterone (ng/dL) 13.3 3.48 12.9 12.9 18.9 Female 18-30 E2 (pg/mL) 122.7 97.4 89.0 89.0 341.0 FSH (mIU/mL) 5.25 3.08 5.15 5.15 9.38 LH (mIU/mL) 5.29 5.82 3.56 3.56 15.3 SHBG (nmol/L) 54.1 28.9 51.2 51.2 107.0 Testosterone (ng/dL) 42.0 16.2 38.2 38.2 74.2 Free Testosterone (ng/dL) 0.59 0.32 0.52 0.52 1.30 29 521 65

(34)

Table 3.

Mean and standard error of natural log-transformed sex hormone-binding globulin (ln-SHBG) across categories of serum PFAS levels in linear regression models (n=540).

ln-SHBG (nmol/L)

Total Male 12–17 Female 12–17 Male 18–30 Female 18–30

No. 540 30 65 180 265 PFOA (ng/ml) <3.63 (<50th) 3.37 (0.07) 3.24 (0.29) 3.50 (0.24)* 3.14 (0.07) 3.83 (0.21) ≤6.78 (50th–75th) 3.43 (0.08) 3.45 (0.29) 3.50 (0.25)* 3.17 (0.09) 3.86 (0.20) ≤9.80 (75th–90th) 3.41 (0.08) 3.67 (0.36) 3.45 (0.29)* 3.20 (0.10) 3.81 (0.22) >9.80 (>90th) 3.33 (0.10) 3.79 (0.39) 2.96 (0.34)* 3.10 (0.14) 3.78 (0.23) PFOS (ng/ml) <5.37 (<25th) 3.47 (0.08) 3.62 (0.29) 3.58 (0.29) 3.13 (0.10) 3.90 (0.21) ≤8.65 (25th–50th) 3.36 (0.08) 3.31 (0.30) 3.36 (0.29) 3.18 (0.10) 3.82 (0.20) ≤13.29 (50th–75th) 3.40 (0.08) 3.47 (0.38) 3.49 (0.24) 3.13 (0.09) 3.89 (0.22) >13.29 (>75th) 3.38 (0.08) 3.46 (0.39) 3.41 (0.44) 3.16 (0.08) 3.80 (0.21) PFNA (ng/ml) <1.64 (<60th) 3.38 (0.07) 3.45 (0.24) 3.46 (0.25) 3.13 (0.08) 3.84 (0.20) ≤6.87 (60th–90th) 3.36 (0.08) 3.51 (0.35) 3.52 (0.26) 3.08 (0.09) 3.79 (0.21) >6.87 (>90th) 3.47 (0.09) 3.59 (0.36) 3.53 (0.41) 3.27 (0.10) 3.92 (0.23) PFUA (ng/ml) <1.53 (<25th) 3.38 (0.08) 3.26 (0.41) 3.81 (0.28) 3.15 (0.09) 3.81 (0.21) ≤6.53 (25th–50th) 3.39 (0.08) 3.62 (0.30) 3.42 (0.32) 3.19 (0.10) 3.80 (0.21) ≤13.41 (50th–75th) 3.40 (0.08) 3.26 (0.37) 3.38 (0.24) 3.17 (0.08) 3.88 (0.21) >13.41 (>75th) 3.40 (0.08) 3.33 (0.36) 3.78 (0.30) 3.12 (0.09) 3.85 (0.20)

Model: adjusted for age, gender, BMI, and high fat diet. PFAS, perfluoroalkyl substances; PFOA, perfluorooctanoic acid; PFOS, perfluorooctane sulfonate; PFNA, perfluorononanoic acid; PFUA, perfluoroundecanoic acid.

*p for trend<0.05

(35)

Table 4.

Mean and standard error of natural log-transformed follicle-stimulating hormone (ln-FSH) across categories of serum PFAS levels in linear regression models (n=540).

ln-FSH (mIU/mL)

Total Male 12–17 Female 12–17 Male 18–30 Female 18–30

No. 540 30 65 180 265 PFOA (ng/ml) <3.63 (<50th) 1.41 (0.08) 1.29 (0.28) 1.47 (0.20) 1.29 (0.08) 1.69 (0.24) ≤6.78 (50th–75th) 1.42 (0.08) 1.58 (0.27) 1.38 (0.21) 1.28 (0.10) 1.65 (0.24) ≤9.80 (75th–90th) 1.34 (0.09) 1.21 (0.34) 1.23 (0.25) 1.23 (0.11) 1.64 (0.25) >9.80 (>90th) 1.46 (0.11) 1.49 (0.36) 1.35 (0.29) 1.13 (0.15) 1.79 (0.26) PFOS (ng/ml) <5.37 (<25th) 1.43 (0.09) 1.50 (0.22)* 1.56 (0.23) 1.20 (0.11) 1.71 (0.25) ≤8.65 (25th–50th) 1.42 (0.08) 1.56 (0.22)* 1.67 (0.23) 1.27 (0.11) 1.66 (0.23) ≤13.29 (50th–75th) 1.42 (0.09) 1.26 (0.28)* 1.36 (0.19) 1.34 (0.10) 1.71 (0.25) >13.29 (>75th) 1.36 (0.08) 0.76 (0.29)* 1.23 (0.35) 1.26 (0.08) 1.69 (0.25) PFNA (ng/ml) <1.64 (<60th) 1.44 (0.08) 1.47 (0.21) 1.53 (0.20) 1.30 (0.08) 1.69 (0.23) ≤6.87 (60th–90th) 1.36 (0.08) 1.43 (0.31) 1.26 (0.22) 1.32 (0.10) 1.58 (0.24) >6.87 (>90th) 1.36 (0.10) 1.19 (0.32) 1.37 (0.33) 1.17 (0.11) 1.73 (0.26) PFUA (ng/ml) <1.53 (<25th) 1.44 (0.08) 1.07 (0.33) 1.59 (0.23)* 1.26 (0.09) 1.73 (0.24) ≤6.53 (25th–50th) 1.41 (0.09) 1.73 (0.24) 1.56 (0.26)* 1.22 (0.11) 1.69 (0.24) ≤13.41 (50th–75th) 1.41 (0.08) 1.45 (0.30) 1.37 (0.20)* 1.33 (0.09) 1.64 (0.24) >13.41 (>75th) 1.35 (0.08) 1.05 (0.29) 1.24 (0.24)* 1.21 (0.10) 1.65 (0.24)

Model: adjusted for age, gender, BMI, and high fat diet. PFAS, perfluoroalkyl substances; PFOA, perfluorooctanoic acid; PFOS, perfluorooctane sulfonate; PFNA, perfluorononanoic acid; PFUA, perfluoroundecanoic acid.

*p for trend<0.05

31 Table 5.

Mean and standard error of natural log-transformed testosterone (ln-T) across categories of serum PFAS levels in linear regression models (n=540).

ln-T (ng/dL)

Total Male 12–17 Female 12–17 Male 18–30 Female 18–30

No. 540 30 65 180 265 PFOA (ng/ml) <3.63 (<50th) 4.96 (0.05) 6.23 (0.23) 3.85 (0.20) 6.32 (0.05) 3.70 (0.15) ≤6.78 (50th–75th) 5.00 (0.06) 5.97 (0.22) 3.96 (0.21) 6.32 (0.06) 3.75 (0.15) ≤9.80 (75th–90th) 4.97 (0.06) 6.46 (0.28) 3.95 (0.25) 6.32 (0.07) 3.65 (0.16) >9.80 (>90th) 4.97 (0.07) 6.48 (0.30) 3.84 (0.29) 6.28 (0.09) 3.71 (0.16) PFOS (ng/ml) <5.37 (<25th) 5.00 (0.06) 6.11 (0.23) 3.97 (0.23)* 6.33 (0.06) 3.73 (0.15) ≤8.65 (25th–50th) 5.02 (0.06) 6.25 (0.24) 4.00 (0.23)* 6.29 (0.07) 3.75 (0.15) ≤13.29 (50th–75th) 4.95 (0.06) 6.24 (0.30) 3.87 (0.19)* 6.32 (0.06) 3.64 (0.16) >13.29 (>75th) 4.94 (0.06) 6.34 (0.31) 3.61 (0.36)* 6.33 (0.05) 3.65 (0.15) PFNA (ng/ml) <1.64 (<60th) 5.00 (0.05) 6.18 (0.19) 3.93 (0.20) 6.33 (0.05) 3.74 (0.15) ≤6.87 (60th–90th) 4.96 (0.06) 6.35 (0.27) 3.85 (0.21) 6.30 (0.06) 3.70 (0.15) >6.87 (>90th) 4.93 (0.06) 6.12 (0.28) 3.64 (0.33) 6.32 (0.06) 3.65 (0.17) PFUA (ng/ml) <1.53 (<25th) 4.97 (0.06) 6.61 (0.29) 4.01 (0.24) 6.30 (0.05) 3.70 (0.15) ≤6.53 (25th–50th) 4.99 (0.06) 5.90 (0.21) 3.95 (0.28) 6.34 (0.06) 3.79 (0.15) ≤13.41 (50th–75th) 5.01 (0.05) 6.33 (0.26) 3.85 (0.21) 6.37 (0.05) 3.78 (0.15) >13.41 (>75th) 4.93 (0.06) 6.53 (0.25) 3.99 (0.26) 6.26 (0.06) 3.69 (0.15)

Model: adjusted for age, gender, BMI, and high fat diet. PFAS,perfluoroalkyl substances; PFOA,

perfluorooctanoic acid; PFOS, perfluorooctane sulfonate; PFNA, perfluorononanoic acid; PFUA, perfluoroundecanoic acid. *p for trend<0.05 523 524 525 526 69

(36)

Table S1.

Linear regression coefficients (95% CI) of log-transformed E2 (pg/mL), FSH (mIU/mL), LH (mIU/mL), testosterone (mIU/mL), free testosterone (mIU/mL), and SHBG (nmol/L) with PFAAs concentrations (ng/mL) in the sample subjects by increase an unit BMI.

Body mass index (kg/m2)

ln-E2 (pg/mL) ln-FSH (mIU/mL) ln-LH (mIU/mL) ln-T (mIU/mL) ln-freeT (mIU/mL) ln-SHBG (nmol/L) PFOA (ng/ml) -0.004(-0.017,0.01) -0.005(-0.017,0.008) -0.01(-0.025,0.006) -0.01(-0.018,0.002) 0.022(-0.012,0.032) -0.063(-0.074,0.052) PFOS (ng/ml) -0.006(-0.02,0.008) -0.004(-0.016,0.008) -0.01(-0.026,0.006) -0.009(-0.018,0.001) 0.023(-0.013,0.033) -0.063(-0.074,0.053) PFNA (ng/ml) -0.004(-0.018,0.01) -0.005(-0.017,0.007) -0.01(-0.026,0.005) -0.01(-0.018,0.002) 0.021(-0.011,0.031) -0.063(-0.073,0.052) PFUA (ng/ml) -0.004(-0.017,0.01) -0.005(-0.017,0.008) -0.01(-0.025,0.006) -0.01(-0.018,0.002) 0.022(-0.012,0.032) -0.063(-0.073,0.052) Model: adjusted for age, gender, and high fat diet.

PFASs, perfluoroalkyl substances; PFOA, perfluorooctanoic acid; PFOS, perfluorooctane sulfonate; PFNA, perfluorononanoic acid; PFUA, perfluoroundecanoic acid.

*p for trend<0.05 Table S2.

Mean and standard error of natural log sex hormone-binding globulin (SHBG) across categories of serum PFAS levels in subpopulation.

log SHBG (nmol/L)

Body mass index (kg/m2) High fat diet

<24 ≥24 No Yes No. 417 123 443 37 PFOA (ng/ml) <3.63 (<50th) 3.56 (0.09) 3.23 (0.13) 3.29 (0.07) 3.61 (0.18) ≤6.78 (50th–75th) 3.62 (0.09) 3.31 (0.15) 3.3 3(0.08) 4.13 (0.18) ≤9.80 (75th–90th) 3.61 (0.10) 3.30 (0.18) 3.34 (0.09) 3.70 (0.20) >9.80 (>90th) 3.45 (0.12) 3.39 (0.21) 3.28 (0.11) 3.54 (0.27) PFOS (ng/ml) <5.37 (<25th) 3.69 (0.09) 3.35 (0.16) 3.39 (0.08) 4.07 (0.29) ≤8.65 (25th–50th) 3.55 (0.09) 3.20 (0.16) 3.26 (0.08) 3.83 (0.23) ≤13.29 (50th–75th) 3.60 (0.09) 3.28 (0.16) 3.32 (0.08) 3.97 (0.23) >13.29 (>75th) 3.52 (0.09) 3.24 (0.15) 3.27 (0.08) 3.72 (0.18) PFNA (ng/ml) <1.64 (<60th) 3.59 (0.08) 3.21 (0.14) 3.32 (0.07) 3.69 (0.17) ≤6.87 (60th–90th) 3.49 (0.09) 3.23 (0.14) 3.27 (0.07) 4.14 (0.20) >6.87 (>90th) 3.63 (0.10) 3.42 (0.19) 3.41 (0.10) 3.79 (0.18) PFUA (ng/ml) <1.53 (<25th) 3.60 (0.09) 3.23 (0.16) 3.31 (0.08) 3.52 (0.23) ≤6.53 (25th–50th) 3.61 (0.09) 3.10 (0.18) 3.28 (0.08) 3.82 (0.19) ≤13.41 (50th–75th) 3.58 (0.09) 3.27 (0.14) 3.30 (0.08) 4.06 (0.19) >13.41 (>75th) 3.55 (0.09) 3.32 (0.15) 3.34 (0.08) 3.62 (0.20)

Model: adjusted for age and gender.

PFAS, perfluoroalkyl substances; PFOA, perfluorooctanoic acid; PFOS, perfluorooctane sulfonate; PFNA, perfluorononanoic acid; PFUA, perfluoroundecanoic acid.

*p for trend<0.05 Table S3.

Mean and standard error of natural log estrogen (E2) across categories of serum PFAS levels in subpopulation.

log E2 (pg/mL)

Body mass index (kg/m2) High fat diet

<24 ≥24 No Yes No. 417 123 443 37 PFOA (ng/ml) <3.63 (<50th) 3.93 (0.12) 4.00 (0.14) 3.96 (0.09) 3.96 (0.26) ≤6.78 (50th–75th) 3.96 (0.12) 3.99 (0.16) 3.98 (0.10) 4.08 (0.27) ≤9.80 (75th–90th) 4.04 (0.13) 3.84 (0.18) 4.05 (0.11) 3.54 (0.30) >9.80 (>90th) 3.93 (0.16) 3.98 (0.21) 3.95 (0.13) 4.12 (0.39) PFOS (ng/ml) <5.37 (<25th) 3.94 (0.13) 3.88 (0.16) 3.93 (0.10) 3.99 (0.38) ≤8.65 (25th–50th) 3.97 (0.12) 3.89 (0.16) 4.00 (0.10) 3.66 (0.30) ≤13.29 (50th–75th) 3.91 (0.12) 4.03 (0.16) 3.95 (0.10) 3.67 (0.31) >13.29 (>75th) 4.03 (0.13) 4.05 (0.15) 4.08 (0.10) 4.05 (0.24) PFNA (ng/ml) <1.64 (<60th) 4.02 (0.11) 3.91 (0.15) 3.97 (0.09) 3.88 (0.24) ≤6.87 (60th–90th) 4.09 (0.13) 4.02 (0.14) 4.07 (0.09) 3.99 (0.30) >6.87 (>90th) 3.75 (0.14) 3.96 (0.19) 3.76 (0.12) 3.84 (0.26) PFUA (ng/ml) <1.53 (<25th) 3.93 (0.12) 3.85 (0.16) 3.93 (0.09) 3.24 (0.30) ≤6.53 (25th–50th) 3.98 (0.13) 3.64 (0.18) 3.91 (0.11) 4.20 (0.24) ≤13.41 (50th–75th) 4.00 (0.12) 4.04 (0.14) 4.02 (0.10) 4.01 (0.24) >13.41 (>75th) 3.97 (0.12) 4.00 (0.15) 4.03 (0.10) 3.69 (0.26)

Model: adjusted for age and gender.

PFAS, perfluoroalkyl substances; PFOA, perfluorooctanoic acid; PFOS, perfluorooctane sulfonate; PFNA, perfluorononanoic acid; PFUA, perfluoroundecanoic acid.

*p for trend<0.05 Table S4.

Mean and standard error of natural log follicle-stimulating hormone (FSH) across categories of serum PFAS levels in subpopulation.

log FSH (mIU/mL)

Body mass index (kg/m2) High fat diet

<24 ≥24 No Yes No. 417 123 443 37 PFOA (ng/ml) <3.63 (<50th) 1.40 (0.09) 1.42 (0.16) 1.44 (0.08) 1.44 (0.23) ≤6.78 (50th–75th) 1.40 (0.10) 1.42 (0.18) 1.45 (0.08) 1.23 (0.24) ≤9.80 (75th–90th) 1.28 (0.11) 1.49 (0.21) 1.36 (0.09) 1.69 (0.27) >9.80 (>90th) 1.45 (0.13) 1.38 (0.24) 1.49 (0.12) 1.37 (0.36) PFOS (ng/ml) <5.37 (<25th) 1.39 (0.10) 1.49 (0.18) 1.45 (0.08) 1.30 (0.35) ≤8.65 (25th–50th) 1.40 (0.10) 1.42 (0.19) 1.43 (0.08) 1.57 (0.28) ≤13.29 (50th–75th) 1.35 (0.10) 1.56 (0.18) 1.45 (0.09) 1.32 (0.28) >13.29 (>75th) 1.36 (0.10) 1.30 (0.17) 1.37 (0.09) 1.42 (0.22) PFNA (ng/ml) <1.64 (<60th) 1.40 (0.09) 1.51 (0.17) 1.45 (0.07) 1.63 (0.20) ≤6.87 (60th–90th) 1.28 (0.10) 1.49 (0.16) 1.38 (0.08) 1.34 (0.25) >6.87 (>90th) 1.38 (0.11) 1.18 (0.22) 1.41 (0.11) 1.23 (0.22) PFUA (ng/ml) <1.53 (<25th) 1.44 (0.1) 1.37 (0.19) 1.47 (0.08) 1.49 (0.30) ≤6.53 (25th–50th) 1.37 (0.1) 1.49 (0.21) 1.41 (0.09) 1.61 (0.24) ≤13.41 (50th–75th) 1.38 (0.1) 1.44 (0.16) 1.43 (0.08) 1.26 (0.25) >13.41 (>75th) 1.30 (0.1) 1.41 (0.18) 1.38 (0.08) 1.33 (0.26)

Model: adjusted for age and gender.

PFAS, perfluoroalkyl substances; PFOA, perfluorooctanoic acid; PFOS, perfluorooctane sulfonate; PFNA, perfluorononanoic acid; PFUA, perfluoroundecanoic acid.

*p for trend<0.05 527 528 529 530 531 532

(37)

33

533

534

參考文獻

相關文件

• The order of nucleotides on a nucleic acid chain specifies the order of amino acids in the primary protein structure. • A sequence of three

S3: And the products were the lipase fatty acid…no, no, fatty acid and glycerol and the enzyme remained unchanged. S1: Our enzyme was amylase and our substrate

Students should also be able to appreciate the interrelation between bonding, structures and properties of substances by learning the properties of metals, giant ionic

Measuring friendship quality in late adolescents and young adults: McGill friendship questionnaires. McGill friendship questionnaire(MFQ- FF) and

PEERS for young adults : Social skills training for adults with autism spectrum disorder and other

gross profit margin, net profit margin, return on capital employed, working capital, current ratio and acid test ratio.. Use accounting ratios to evaluate a company’s profitability

In the citric acid cycle, how many molecules of FADH are produced per molecule of glucose.. 111; moderate;

This database includes antigen’s PDB_ID, all sites (include interaction and non-interaction) of a nine amino acid sequence of primary structure and secondary structure.. After