Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Science of the Total Environment 625 (2018) 566–574

Contents lists available at ScienceDirect

Science of the Total Environment

journal homepage: www.elsevier.com/locate/scitotenv

PFOA is associated with diabetes and metabolic alteration in US men:


National Health and Nutrition Examination Survey 2003–2012
Xiaowei He a,1, Yuanxin Liu a,1, Bo Xu b, Liubao Gu a, Wei Tang a,⁎
a
Department of Endocrinology, Islet Cell Senescense and Function Research Laboratory, Jiangsu Province Geriatric Institute, 30 Luojia Road, Nanjing, Jiangsu 210024, China
b
School of Public Health, Nanjing Medical University, Nanjing 211166, China

H I G H L I G H T S G R A P H I C A L A B S T R A C T

• 7904 adults were used to examine asso-


ciations of PFAS with diabetes and me-
tabolite levels.
• Serum PFOA was positively associated
with diabetes mellitus in men.
• PFOA may disrupt cholesterol metabo-
lism at environmentally exposures.

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

Article history: Exposure to perfluoroalkyl substances (PFAS) is associated with a range of adverse health effects. However, it re-
Received 24 September 2017 mains unclear whether PFAS at environmentally relevant exposure levels are related to diabetes and metabolite con-
Received in revised form 16 December 2017 centrations in adults. Using cross-sectional data from 7904 adults (age ≥ 20 years) in the 2003–2012 National Health
Accepted 17 December 2017
and Nutrition Examination Survey (NHANES), we examined the association of PFAS with the prevalence of diabetes
Available online xxxx
and metabolite concentrations. A multivariate logistic regression was applied to investigate the associations of dia-
Editor: D. Barcelo betes prevalence with serum perfluorooctanoate (PFOA), perfluorooctane sulfonate (PFOS), perfluorohexane sulfo-
nate (PFHxS) and perfluorononanoate (PFNA) levels. A multivariate generalised linear regression was further
Keywords: performed to investigate the associations between PFAS exposure and some metabolites. We identified a strong pos-
Perfluoroalkyl substances itive association between serum PFOA and diabetes prevalence in men with an adjusted model (OR: 2.66, 95% CI:
Perfluorooctanoate 1.63–4.35; P for trend = 0.001). No significant association between serum PFOA and diabetes prevalence was ob-
Diabetes served in women (OR: 1.47, 95% CI: 0.88–2.46; P for trend = 0.737). Furthermore, diabetes was not related to
Metabolites PFOS, PFHxS and PFNA, regardless of gender. In the gender-stratified generalised linear models, men and women
Total cholesterol
with the highest PFOA levels demonstrated a 1.43% (95% CI: 0.62%–2.34%) and a 1.07% (95% CI: 0.27%–1.97%) greater
increase in serum total cholesterol (P for trend = 0.006 and 0.001) compared to those with the lowest PFOA levels.
There were no significant associations between serum PFOA and other metabolites. These results provide epidemi-
ological evidence that environment-related levels of serum PFOA may be positively associated with the prevalence
of diabetes in men and with total cholesterol in adults. Further clinical and animal studies are urgently needed to
elucidate putative causal relationships and shed light on the potential mode of action involved.
© 2017 Published by Elsevier B.V.

⁎ Corresponding author.
E-mail address: drtangwei@aliyun.com (W. Tang).
1
These authors contributed equally to this work.

https://doi.org/10.1016/j.scitotenv.2017.12.186
0048-9697/© 2017 Published by Elsevier B.V.
X. He et al. / Science of the Total Environment 625 (2018) 566–574 567

1. Introduction contaminate various environmental media (Butt et al., 2010). Mean-


while, biomonitoring of the general population has shown widespread
Diabetes mellitus, usually referred to as diabetes, is a group of meta- exposure to certain PFAS (Christensen et al., 2016). Of note, mean
bolic disorders characterised by high blood glucose levels over a serum half-lives in humans are estimated to be 4.8 years for PFOS,
prolonged period (Moyer et al., 2012). Without effective management, 3.5 years for perfluorooctanoate (PFOA) (Olsen et al., 2007) and
diabetes can induce many health complications. Recent estimates attri- 8.5 years for perfluorohexane sulfonate (PFHxS). Once absorbed, these
bute almost 5.1 million deaths to diabetes in adults, representing 8.4% of chemicals can persist in the body by binding to liver and serum proteins
all-cause deaths globally (IDF Diabetes Atlas Group, 2015). Risk factors (Ng and Hungerbuehler, 2015). Furthermore, the widely utilised newer
for diabetes include genetic predisposition, ethnic background, age, "replacement" of PFAS ensures a sustained exposure risk (Wang et al.,
lack of physical exercise, obesity and use of tobacco (Karnes et al., 2013).
2014; Thiering et al., 2011). Recent research has indicated that environ- High serum PFOA level was found among workers with occupational
mental exposure could have contributed to the recent dramatic surge in exposure to PFOA at a DuPont chemical plant (Steenland and Woskie,
diabetes (Bond and Dietrich, 2017; Honda et al., 2017). Environmental 2012). Workers exposed to PFOA showed a twofold increase in diabetes
chemicals used in most of today's family households may have mortality in comparison to other non-exposed regional workers
endocrine-disrupting properties and thus disrupt the endogenous hor- (Leonard et al., 2008; Steenland and Woskie, 2012). However, studies
mone system (Audouze et al., 2013) (Gore et al., 2015). These chemicals of residents in this community indicate a null association between
may cause impaired insulin secretion and insulin sensitivity, causing the PFOA exposure and type II diabetes, both cross-sectionally and prospec-
development of diabetes (Domazet et al., 2016). tively (Karnes et al., 2014; MacNeil et al., 2009). Notably, exposure
Perfluoroalkyl substances (PFAS) are human-made compounds that levels in these workers or residents are much higher than in general
have been used in a multitude of industrial and commercial applica- US populations of non-exposed region.
tions, including fire extinguishing foams, photographic emulsifiers and As a review by Post et al. mentions (Post et al., 2012), this study's
stain-, grease- and water-resistant protection for clothing, furniture, dose-response curve presented steeper slopes at low levels of PFOA ex-
carpet, leather, and paper (Calafat et al., 2007; Lau et al., 2007). PFAS posure than at high levels of PFOA exposure. This suggests the great im-
contain a group of substances that are classified as persistent organic portance of exploring low-dose effects in the general population.
pollutants due to their strong resistance to biodegeneration, hydrolysis, Nevertheless, the role of PFOA in prevalent diabetes is still largely un-
photolysis and atmospheric photooxidation (Domazet et al., 2016). The known; and inconsistent relationships were observed in the studies
production of perfluorooctane sulfonate (PFOS) and some other PFAS on the general population.
has been voluntarily discontinued in the United States (Buck et al., To further clarify the relationship between PFAS and diabetes prev-
2011). However, due to its ubiquity and persistence, PFAS still alence, we performed a cross-sectional analysis of the 2003–2012

Fig. 1. Flow chart depicting the final analysed samples for each outcome from five successive waves of NHANES cycle (2003–2012). NHANES, National Health and Nutrition Examination
Survey; PFAS, perfluoroalkyl substances; FPG, fasting plasma glucose; Fins, Fasting insulin; HOMA-IR, homeostasis model assessment of insulin resistance; TC, total cholesterol; HDL-C,
HDL-cholesterol; LDL-C, LDL-cholesterol; TG, triglycerides.
568 X. He et al. / Science of the Total Environment 625 (2018) 566–574

National Health and Nutrition Examination Survey (NHANES) to exam- were included in this analysis. Concentrations below the limit of detec-
ine the association of serum PFAS levels with diagnosed diabetes in tion (LOD) were reported by NHANES as the LOD divided by the square
adults, stratified by gender. root of two.

2. Research design and methods 2.5. Potential confounders

2.1. Study population The following variables were chosen a priori as covariates in the
models because of their biological relevance to the outcomes: age,
Publicly available data for participants who enrolled in this study race, body mass index (BMI), education level, energy intake, serum co-
were merged from five cycles of the NHANES (2003–2004, tinine (a exposure marker of environmental cigarette smoke), alcohol
2005–2006, 2007–2008, 2009–2010, and 2011–2012). Detailed infor- consumption, poverty-income ratio (PIR; family income divided by
mation of the survey design and methods are available on the the federal poverty threshold controlling for inflation and family size),
NHANES website [Centers for Disease Control and Prevention (CDC)]. and time spent in front of a screen (watching television, playing video
Briefly, the NHANES is a series of continuous, multicomponent cross- games and using computers; measured as hours per day in the forego-
sectional surveys designed to be nationally representative of the non- ing 30 days) (Goldfield et al., 2013; Holst et al., 2017; Jarvandi et al.,
institutionalised U.S. civilian population. After providing written in- 2015). The information of these variables was extracted from the
formed consent, each participant visited a mobile examination center NHANES questionnaires. In addition, a variable was considered as a con-
(MEC) for physical assessment, examination, and biological sample founder if its inclusion in the model could change the beta coefficient for
collection. any quartiles of the PFAS exposure by N 10% (James-Todd et al., 2012).
Individuals participating in the NHANES over a range of 10 years
were selected to from a random representative subset for the measure- 2.6. Statistical analysis
ment of serum PFAS and serum metabolite levels. Of 24,146 adults
≥20 years of age who engaged in both the NHANES interview and the Serum concentrations of PFAS were categorized in quartiles on the
examination, those who had missing PFAS information or were current- basis of the sample distribution. We applied a multivariate logistic
ly pregnant were excluded. In a final sample of 7904 participants, we in-
vestigated the association of serum PFAS levels with diabetes Table 1
prevalence and metabolite concentrations. Due to the self-reported na- Characteristics of 2003–2012 NHANES study participants and distribution of serum
ture of this assessment in NHANES, we cannot identify the type of diabe- perfluoroalkyl substances (PFAS) by gender.a

tes. However, it must be pointed out that the vast majority of Characteristics Male Female
participants with diabetes in NHANES are likely to be type II in origin, (n = 3956) (n = 3948)
since this is a nationally general population study among adults in US Age (years) 50.22 ± 0.29 49.33 ± 0.29
(Bainbridge et al., 2008; Menke et al., 2013). Final analysed samples BMI (kg/m2) 28.45 ± 0.10 29.22 ± 0.12
for each outcome among eligible populations are summarised in Fig. 1. Energy intake (kcal) 2375.66 ± 15.23 1744.19 ± 10.40
Serum cotinine (ng/ml) 71.77 ± 2.25 42.41 ± 1.69
Race (%)
2.2. Primary outcome: diabetes mellitus Mexican American 16.8 17.0
Other Hispanic 7.2 8.6
Participants were asked by NHANES interviewers if they had ever Non-Hispanic White 48.4 47.2
been told by a doctor or other health professional that they had diabetes Non-Hispanic Black 20.6 20.4
Other race - including multi-racial 7.1 6.8
or sugar diabetes (other than during pregnancy). Based on answering Education level (%)
“yes” to this question, participants were coded as having been diag- Less than 9th grade 13.9 11.8
nosed with diabetes. 9–11th grade 15.3 15.5
(includes 12th grade with no diploma)
High school grad/GED or equivalent 23.3 22.5
2.3. Secondary outcome: metabolite concentrations
Some college or AA degree 26.0 29.6
College graduate or above 21.2 20.4
To observe whether there are relationships between PFAS and me- Time spent in front of a screen (%)
tabolism alteration, we conducted additional analyses in a random sub- ≤2 h 41.9 45.3
group with metabolite data. We explored the associations between N2 h 58.1 54.7
PIR (%)
PFAS with markers of glucose metabolism [fasting plasma glucose ≤1 17.1 20.2
(FPG), fasting insulin levels (Fins), the homeostasis model assessment N1 75.0 72.0
of insulin resistance (HOMA-IR)] and lipid metabolism [total cholesterol Alcohol usage (%)
(TC), triglycerides (TG), high-density lipoprotein cholesterol (HDL-C), Yes 76.9 53.4
No 15.6 36.7
low-density lipoprotein cholesterol (LDL-C)] and blood pressure. Ab-
BMI (%)
normalities of these metabolic parameters are known to increase the b25 28.4 31.5
risk for diabetes (Lorenzo et al., 2007; Z. Zhang et al., 2014). 25–30 38.8 28.7
Detailed information on sample collection and treatment at the U.S. N30 31.4 38.3
CDC are available in the previous studies (Peng et al., 2015; Xu et al., PFOA (ng/ml) 4.50 ± 0.06 3.46 ± 0.04
PFOS (ng/ml) 20.80 ± 0.32 14.51 ± 0.26
2015).
PFHxS (ng/ml) 2.88 ± 0.05 1.94 ± 0.04
PFNA (ng/ml) 1.52 ± 0.02 1.30 ± 0.03
2.4. Primary exposure: serum PFAS levels
Age, BMI, energy intake, serum cotinine and PFAS are presented as mean values and their
standard errors.
Briefly, serum PFAS levels were analysed at the U.S. CDC using online Race, education level, time spent in front of a screen, alcohol usage and BMI are presented
solid-phase extraction–high performance liquid chromatography– as percentages.
turbo ion spray-tandem mass spectrometry (SPE-HPLC-TIS-MS/MS), BMI, body mass index; PIR, poverty-Income ratio; PFOA, perfluorooctanoic acid;
PFOS, perfluorooctane sulfonic acid; PFHxS, perfluorohexane sulfonic acid; PFNA,
as reported previously (Calafat et al., 2007; Jain, 2014). perfluorononanoic acid.
Serum concentrations of four PFAS (PFOA, PFOS, PFHxS, and PFNA), a
Subjects were excluded if they had a positive laboratory pregnancy test or self-re-
which were detectable in N 98% of the 2003–2012 survey participants, ported to be pregnant at exam (n = 202).
X. He et al. / Science of the Total Environment 625 (2018) 566–574 569

Table 2 (IBM Corp., Armonk, NY, USA) was applied for all statistical analyses.
Distribution of diabetes outcome and concentrations of serum metabolites in the A P value b 0.05 was designated as the criteria for statistical significance.
participants.

Characteristics Male Female 3. Results


n Mean ± SE (%) n Mean ± SE (%)
3.1. Baseline characteristics
Diabetes (%) 3874 11.8 3865 11.6
FPG (mg/dL) 1918 111.30 ± 0.87 1928 104.81 ± 0.77
Fins (uU/mL) 1909 13.98 ± 0.47 1917 12.50 ± 0.27 Table 1 shows the general characteristics and mean concentrations
HOMA-IR 1907 4.12 ± 0.17 1914 3.41 ± 0.09 of serum PFAS levels among the participants included in the current
TC (mg/dL) 3954 193.5 ± 0.67 3947 200.47 ± 0.66 study from the NHANES 2003–2012 database. Table 2 presents the dis-
HDL-C (mg/dL) 3954 48.32 ± 0.23 3946 57.86 ± 0.26
LDL-C (mg/dL) 1844 115.28 ± 0.84 1903 116.70 ± 0.83
tribution of the diabetes outcome and serum metabolite levels of the
TG (mg/dL) 1916 148.58 ± 3.00 1927 124.47 ± 2.34 participants. Diabetes mellitus was reported in 11.8% of men and
SBP (mm Hg) 3804 125.15 ± 0.28 3739 123.44 ± 0.35 11.6% of women in this study population.
DBP (mm Hg) 3804 71.18 ± 0.21 3739 68.73 ± 0.22

FPG, fasting plasma glucose; Fins, fasting insulin; HOMA-IR, homeostasis model assessment 3.2. Diabetes mellitus
of insulin resistance; TC, total cholesterol; HDL-C, HDL-cholesterol; LDL-C, LDL-cholester-
ol; TG, triglyceride; SBP, systolic blood pressure; DBP, diastolic blood pressure.
The primary effect estimates from the multinomial logistic regres-
sion models adjusted for a group of covariates for individual and cumu-
regression analysis to investigate the association of a diabetes outcome lative PFAS are presented in Table 3. Notably, significant positive
with each PFAS separately. As gender is a particularly crucial associations were observed between diabetes prevalence and quartile
predicator, we conducted a separate analysis stratified by gender. 2 (Q2; OR: 2.13, 95% CI: 1.30, 3.46), Q3 (OR: 2.44, 95% CI: 1.49, 3.98)
To evaluate the additive effect of four PFAS, we categorized each and Q4 (OR: 2.67, 95% CI: 1.63, 4.38) of PFOA, relative to the lowest
PFAS into 10 groups using 10th percentiles. The cumulative PFAS Q1 of exposure (P for trend = 0.001) in men. Additionally, there was
level was made by adding up the category number of each PFAS, a slight association between diabetes prevalence and each quartile of
resulting in a value of 4–40. Then the cumulative PFAS was catego- PFOA in women, although the dose-response relationship in women
rized into quartiles, making four groups (Moon, 2014). We showed was less strong and clear than in men (Fig. 2A). Furthermore, diabetes
effect estimates and their matching 95% confidence intervals (CIs) prevalence was not significantly associated with any of the categorical
for each quartile compared with the lowest quartile (the reference PFOS, PFHxS, PFNA, or cumulative PFAS exposure, regardless of gender.
group). P for trend was performed by considering the PFAS category
as a linear variable in the models. 3.3. Metabolism alteration
A generalised linear model was performed to estimate the associa-
tions between increases in the interquartile ratio (IQ ratio = 75th/ Because we found a significant association between PFOA exposure
25th percentiles of PFAS level) for levels of metabolites and PFAS. Con- and the prevalence of diabetes, we further explored the association be-
tinuous outcomes were log-transformed to approximate a normal dis- tween some serum metabolites and PFOA exposure. Estimated % diff
tribution. Gender was used as the stratified variable to conduct a and 95% CI in serum TC concentrations for each IQ ratio increase in
separate analysis. Testing for linear trends was performed by entering serum PFOA levels are presented separately for men and women in
the PFAS quartiles as a continuous variable into the same model. The Table 4 and Fig. 2B. Men with the highest PFOA levels demonstrated a
magnitudes of association were presented as the average percentage 1.43% (95% CI: 0.62%, 2.34%) greater increase in serum TC (P for trend
difference (% diff) in metabolic levels for comparison of interquartile ra- b 0.001) than participants with the lowest serum PFOA levels. Women
tios (IQ ratio) for PFOA, calculated as [(IQ ratiobeta) − 1] × 100. This in Q3 and Q4 of PFOA levels showed a 1.16% (95% CI: 0.44%, 1.97%)
method scales the magnitudes of association to an exposure range, and 1.07% (95% CI: 0.27%, 1.97%) greater increase in serum TC (P for
which existed in both genders of the study population. trend = 0.001) than those participants in Q1 of PFOA levels. We found
All regression models were conducted after adjusting for age, race, no association between PFOA concentration and FPG, Fins, HOMA-IR,
BMI, education levels, energy intake, serum cotinine, alcohol consump- HDL-C, LDL-C, SBP or DBP (Supplementary materials). The relationships
tion, PIR, and the time in front of a screen (watching television, playing between the biomarkers and PFOS, PFHxS or PFNA are presented in the
video games or using computers). The IBM SPSS software, version 20.0 Supplementary materials.

Table 3
Odd ratio (OR) for the associations between the quartiles of serum PFAS and diabetes prevalence.

PFAS Gender Q1 Q2 Q3 Q4 P for trend

PFOA Male Reference 2.13a (1.30, 3.46) 2.44a (1.49, 3.98) 2.67a (1.63, 4.38) 0.001
PFOS Reference 1.32 (0.71, 2.45) 1.64 (0.92, 2.93) 1.75 (1.00, 3.04) 0.305
PFHxS Reference 1.99 (1.19, 3.33) 1.87 (1.15, 3.05) 2.31 (1.37, 3.91) 0.071
PFNA Reference 1.25 (0.77, 2.04) 1.17 (0.74, 1.87) 1.19 (0.73, 1.95) 0.457
Cumulative PFAS Reference 1.18 (0.84, 1.65) 1.34 (0.97, 1.86) 1.55 (1.11, 2.14) 0.363
PFOA Female Reference 1.38 (0.87, 2.19) 1.57 (0.95, 2.61) 1.47 (0.87, 2.48) 0.737
PFOS Reference 1.11 (0.66, 1.88) 1.06 (0.63, 1.78) 1.41 (0.82, 2.41) 0.829
PFHxS Reference 0.65 (0.41, 1.03) 0.87 (0.52, 1.43) 1.22 (0.71, 2.11) 0.056
PFNA Reference 0.98 (0.63, 1.54) 1.50 (0.88, 2.57) 1.01 (0.62, 1.65) 0.250
Cumulative PFAS Reference 1.02 (0.75, 1.38) 1.17 (0.84, 1.63) 1.13 (0.80, 1.59) 0.162

Multinomial logistic regression models were used, adjusted for age, race, BMI, education level, energy intake, serum cotinine, alcohol consumption, PIR and the time in front of TV, video
game and computer.
Quartiles (Q) of PFOA (ng/ml): Q1: b2.1; Q2: 2.1–3.34; Q3: 3.34–5.1; Q4: N5.1.
Quartiles (Q) of PFOS (ng/ml): Q1: b7.305; Q2: 7.305–13.1; Q3: 13.1–22.5; Q4: N22.5.
Quartiles (Q) of PFHxS (ng/ml): Q1: b0.9; Q2: 0.9–1.64; Q3: 1.64–2.9; Q4: N2.9.
Quartiles (Q) of PFNA (ng/ml): Q1: b0.738; Q2: 0.738–1.07; Q3: 1.07–1.64; Q4: N1.64.
PFAS, perfluoroalkyl substances; PFOA, perfluorooctanoic acid; PFOS, perfluorooctane sulfonic acid; PFHxS, perfluorohexane sulfonic acid; PFNA, perfluorononanoic acid.
a
P b 0.05.
570 X. He et al. / Science of the Total Environment 625 (2018) 566–574

Fig. 2. Adjusted odd ratio (OR) and 95% confidence intervals (95% CI) for diabetes risk in U.S. adults 2003–2012 by increasing quartiles of serum PFOA levels by gender (A). Estimated
percent difference (% diff) and 95% CI in serum total cholesterol (TC) concentrations in U.S. adults 2003–2012 for each interquartile ratio (IQ ratio) increase in serum PFOA levels by
gender (B). Quartiles (Q) of PFOA (ng/ml): Q1: b2.1; Q2: 2.1–3.34; Q3: 3.34–5.1; Q4: N5.1.

To compare with a previous study demonstrating the association be- diabetes prevalence in adults. To our knowledge, this is the first study
tween PFOA levels and TC in a middle-aged population (Eriksen et al., to uncover this difference in adults. This finding may be attributed to
2013), we conducted an effect estimation to examine the relationship the divergent pharmacokinetics of PFOA between genders. In a recent
between PFOA and TC, stratified by age and gender. As presented in study of PFOA exposure in rats, female rats excreted approximately
Table 5, strong associations were observed in men aged 40–50 (% diff: 60% or more of a PFOA dose in urine and faeces within 24 h, whereas
2.88%, 95% CI: 0.89%, 4.91%) and 50–60 (% diff: 2.52%, 95% CI: 0.44%, male rats excreted only 9.0% of the same PFOA dose in urine within a
4.73%), as well as women aged 60–70 (% diff: 2.88%, 95% CI: 0.80%, prolonged period of 12 days (Kim et al., 2016). Previous studies have
5.00%). Scatter plots and fitted lines with 95% CIs of the associations be- also suggested that gender difference for the effect of PFOA exposure
tween PFOA concentrations and serum TC are shown in Fig. 3 for men could be partially explained by the specific excretion pathway of fe-
aged 40–60 (Fig. 3A) and women aged 60–70 (Fig. 3B). males, such as menstruation, delivery and/or lactation (Li et al., 2017;
Y. Zhang et al., 2014). In addition, PFOA is one of the potential endocrine
4. Discussion disrupting chemicals (Kjeldsen and Bonefeld-Jorgensen, 2013) and it
can interact with estrogen receptor α (Buhrke et al., 2015; Kang et al.,
Using data from the representative NHANES samples allowed us to 2016).
explore the associations between four PFAS and the prevalence of dia- In our study, we examined the associations between the serum PFOA
betes, as well as metabolite concentrations. The most prominent find- levels and glucose metabolism markers, comprising FPG, Fins and
ings were for TC in the overall population and diabetes in men. HOMA-IR in adults. Consistent with some previous studies (Domazet
A cross-sectional study in an elder Swedish population demonstrat- et al., 2016; Fisher et al., 2013; Karnes et al., 2014; Lin et al., 2009;
ed no significant association between PFOA [median: 3.3, interquartile Lind et al., 2014; MacNeil et al., 2009; Nelson et al., 2010), none of
range (IQR): 2.5, 4.4 ng/ml] and diabetes prevalence (Lind et al., these analysed parameters were related to PFOA exposure, although
2014). However, in a recent study conducted in 571 working-age small number of population was reported in these studies. Besides
Taiwanese participants, PFOA (median: 8.0, IQR: 5.8, 10.5 ng/ml) was these analysed parameters above, beta cell is central to the develop-
shown to be inversely associated with the risk of diabetes (Su et al., ment and progression of diabetes (Cnop et al., 2005; Halban et al.,
2016). In our study, an adverse impact effect of PFOA (median: 3.3, 2014). Previous studies have suggested the relationship between
IQR: 2.1, 5.1 ng/ml) against diabetes risk was demonstrated. The dis- PFOA and beta cell function. In a multicenter prospective investigation,
crepancy between our discovery and the results from previous studies childhood PFOA exposure could predict impaired beta cell function at
may be due to divergent criteria used to define diabetes, different expo- 15 years of age (Domazet et al., 2016). However, in a NHANES study ex-
sure levels of PFAS, different sampling or analysing procedures. In an- ploring the role of PFOA/PFOS isomers in glucose homeostasis, increased
other study conducted with 258 pregnant women, PFOA (median: linear PFOA was associated with an enhancement of beta cell function
3.3 ng/ml) was significantly associated with an increased GDM risk (Liu et al., 2017). In addition, strong positive association between
(Zhang et al., 2015). In the present study, we discovered a significant serum PFOA concentrations and β-cell function was observed in 969
gender difference for the associations between PFOA exposure and adults from the NHANES 1999–2004 (Lin et al., 2009). Thus, the exact

Table 4
Estimated percent difference (% diff) and 95% confidence intervals (95% CI) in serum total cholesterol (TC) concentrations in US adults 2003–2012 for each interquartile ratio (IQ ratio)
increase in serum PFOA levels.

TC Males Females

% diff 95% CI P for trend % diff 95% CI P for trend

Q1 Reference b0.001 Reference 0.001


Q2 0.44% −0.44%, 1.34% 0.53% −0.18%, 1.25%
Q3 0.80% −0.09%, 1.70% 1.16%a 0.44%, 1.97%
Q4 1.43%a 0.62%, 2.34% 1.07%a 0.27%, 1.97%

TC was Log-transformed in models.


% diff = [(IQ ratiobeta) − 1] ∗ 100.
IQ ratio = 75th/25th percentiles of serum PFOA: 2.43.
Quartiles (Q) of PFOA (ng/ml): Q1: b2.1; Q2: 2.1–3.34; Q3: 3.34–5.1; Q4: N5.1.
PFOA, perfluorooctanoic acid.
a
P b 0.05.
X. He et al. / Science of the Total Environment 625 (2018) 566–574 571

mechanism that links PFOA to glucose metabolism is still unknown and 2006). More epidemiological and mechanistic studies are needed to
mixed. verify the age difference of PFOA exposure effect and to elucidate the
In contrast, the evidence for a relationship between PFOA exposure potential reasons.
and lipid metabolism is much stronger. Although results in humans The underlying molecular mechanism for the association between
are not entirely consistent, the general trend is one of positive associa- PFOA and diabetes has yet to be elucidated. It has been reported that
tions between PFOA concentration and TC levels, mainly in occupational PFAS can bind to peroxisome proliferators activated receptors (PPARs)
workers (Costa et al., 2009; Sakr et al., 2007) or in population of com- (Lau et al., 2007). PPARα is mainly expressed in the liver and is involved
munities surrounding the DuPont chemical plant (Steenland et al., in lipid homeostasis, fatty acid catabolism and inflammation (Wolf et al.,
2009). Notably, we found that women in Q3 of PFOA levels showed a 2008). PPARγ activation has been shown to influence insulin resistance
greater increase in serum TC than those participants in Q4 of PFOA and insulin secretion (Lebovitz and Banerji, 2001; Lupi et al., 2004). The
levels. Thus, one should be cautious about concluding that PFOA may fibrate class of cholesterol-lowering medications (one of the PPARα li-
have a relationship with TC in women. But interestingly, the significant gands) may inhibit secretion of cholesterol from the liver, resulting in
association was only observed in women aged over 70. This age differ- reduced cholesterol in the serum (Kennedy et al., 2004). However, our
ence may be the reason for the disappearance of PFOA dose response results are not completely consistent with prior animal findings, sug-
in women. The aforementioned prospective study conducted in Danish gesting PFOA might exert its effects via alternative or even multiple
individuals found an association between PFOA and high TC levels in a pathways. Furthermore, interspecies differences may partially explain
subset of participants 50–60 years old (Eriksen et al., 2013). However, the inconsistency of cholesterol findings between animal and human
its population was limited to 50–60 years old and difference between studies.
genders was not analysed. Two previous studies using NHANES data A recent epidemiological study conducted in a Chinese adult male
also reported a relationship between serum PFOA and lipid metabolism cohort has suggested that low-level environmental exposure to PFAS
among general US adults (Liu et al., 2017; Nelson et al., 2010). However, was related to oxidative stress in humans and increased the risk of dia-
the results from these two study were different from ours. Specifically, betes (Wang et al., 2017). An in vitro study by Suh et al. discovered that
Nelson et al. found that associations were identical in each age and PFOA exposure was linked to increased oxidative stress and mitochon-
sex subgroups with greater magnitude among persons 60–80 years of drial dysfunction, inducing apoptosis in pancreatic β-cells (Suh et al.,
age. In the study conducted by Liu et al., although increased linear 2017). Additionally, PFOA can cause metabolome disorders involving
PFOA was associated with increases in TC, age and sex subgroups pollutant detoxification, antioxidation and nitric oxide signalling path-
were not implemented. In a study conducted in post-weaning rats, ways. Furthermore, Yan et al. found that the serine/threonine protein
age effect on PFOA plasma concentration has been investigated. A kinase (AKT) signalling pathway was activated in PFOA exposure (Yan
slightly but significantly higher plasma concentration occurred in et al., 2015). These results indicate that PPAR-independent mechanisms
older males while significantly lower (approximately 10-fold) plasma may exist for PFOA-related diabetogenesis and further efforts should be
PFOA concentrations was observed in older females (Hinderliter et al., made to establish the underlying molecular mechanism.

Table 5
Estimated percent difference (% diff) and 95% confidence intervals (95% CI) in serum total cholesterol (TC) concentrations in US adults 2003–2012 for each interquartile ratio (IQ ratio)
increase in serum PFOA levels stratified by age.

Age (years) TC Males Females

% diff 95% CI P for trend % diff 95% CI P for trend

20–30 Q1 Reference 0.111 Reference 0.226


Q2 0.62% −1.41%, 2.70% 1.88% 0.44%, 3.34%
Q3 −0.18% −2.10%, 1.79% 1.61% 0.00%, 3.25%
Q4 1.61% −0.27%, 3.62% −0.18% −2.20%, 1.88%
30–40 Q1 Reference 0.438 Reference 0.190
Q2 0.71% −1.32%, 2.70% −0.09% −1.67%, 1.43%
Q3 0.71% −1.15%, 2.60% 1.97% 0.27%, 3.80%
Q4 0.98% −0.88%, 2.88% 1.16% −1.06%, 3.43%
40–50 Q1 Reference 0.004 Reference 0.001
Q2 1.52% −0.62%, 3.71% 2.43% 0.89%, 4.08%
Q3 1.16% −0.88%, 3.25% 3.25% 1.43%, 5.00%
Q4 2.88%a 0.89%, 4.91% 1.43% −0.44%, 3.43%
50–60 Q1 Reference 0.002 Reference 0.610
Q2 0.53% −1.59%, 2.79% 0.71% −1.15%, 2.70%
Q3 2.24% 0.09%, 4.45% 0.80% −1.24%, 2.88%
Q4 2.52%a 0.44%, 4.73% 2.88% 0.80%, 5.00%
60–70 Q1 Reference 0.341 Reference 0.006
Q2 0.44% −1.67%, 2.61% −0.35% −2.37%, 1.79%
Q3 0.62% −1.59%, 2.79% 0.80% −1.24%, 2.88%
Q4 1.25% −0.88%, 3.34% 2.88%a 0.80%, 5.00%
70–80 Q1 Reference 0.453 Reference 0.965
Q2 −0.88% −3.06%, 1.43% −1.15% −3.32%, 1.16%
Q3 0.98% −1.32%, 3.34% −0.27% −2.46%, 1.97%
Q4 0.36% −1.85%, 2.61% 0.36% −1.76%, 2.43%
N80 Q1 Reference 0.466 Reference 0.591
Q2 1.34% −4.68%, 7.84% −4.51% −11.14%, 2.61%
Q3 −2.63% −8.33%, 3.34% 0.00% −7.35%, 7.84%
Q4 −1.24% −6.61%, 4.45% −1.41% −8.74%, 6.60%

TC was Log-transformed in model.


% diff = [(IQ ratiobeta) − 1] ∗ 100.
IQ ratio = 75th/25th percentiles of serum PFOA: 2.43.
Quartiles (Q) of PFOA (ng/ml): Q1: b2.1; Q2: 2.1–3.34; Q3: 3.34–5.1; Q4: N5.1.
PFOA, perfluorooctanoic acid.
a
P b 0.05.
572 X. He et al. / Science of the Total Environment 625 (2018) 566–574

PFOA has been determined worldwide in human blood. The out separate analyses stratified by age and sex, which can explore the
median concentration of PFOA in adult blood from Queensland, sensitivity of different groups to these chemicals. Our study has several
Australia (6.4 ng/ml) (Toms et al., 2009) and Shenyang, China limitations. We cannot rule out the possibility of reverse causation be-
(6.19 ng/ml) (Bao et al., 2017) were higher than those reported for cause of the cross-sectional study. In general, PFAS can be excreted
adults from other regions (b 0.5–1.61 ng/ml) (i.e., Korea, arctic through the kidneys. The lower PFOA filtration rate in kidney will lead
Russia, Uzbekistan and Afghanistan) (Cho et al., 2015; Hanssen to higher serum concentrations of PFAS. A Previous study has shown
et al., 2013; Hemat et al., 2010). It should be noted that the exposure that PFAS filtration rate was lower in diabetic patients than in normal
of PFOA tended to deviate from the normal distribution. In the stud- subjects (Conway et al., 2016). As such, it is possible that some of
ies of Rocca et al. conducted in Italy, a dispersed distribution of PFOA these associations are due to reduced PFAS excretion among patients
was observed, with a large fraction of samples at or below LOD and with diabetes. Moreover, a lot of environmental chemicals (such as
part of samples at high levels (La Rocca et al., 2015; La Rocca et al., polycyclic aromatic hydrocarbons (Alshaarawy et al., 2014), bisphenol
2014). Even though the background concentration of PFOA is lower A (Ahmadkhaniha et al., 2014), phthalates (Mamtani et al., 2016), mer-
now than it was a decade ago, its long half-life could suggest the ef- cury (He et al., 2013), pesticide (Saldana et al., 2007) and arsenic (Rhee
fect of bioaccumulation, leading to long-term body burdens and et al., 2013)) were potentially associated with the risk of diabetes. These
health risks (Poet et al., 2016). Humans can be exposed to PFOA chemicals were not evaluated in our analysis, which may have an im-
through various routes, such as diet, drinking water, inhalation and pact on the association between PFOA and diabetes. Future studies
ingestion of indoor dust (Ferguson et al., 2013). Other than plasma may need to assess interaction effect of different chemicals on the risk
and tissues, PFOA can also exist in umbilical cord blood (Arbuckle of diabetes.
et al., 2013) and breast milk (Fromme et al., 2010). Thus, further ef-
forts are still needed to limit emissions of PFOA.
This study has some critical strengths. While many previous investi- 5. Conclusion
gations of PFOA health effects were based on small samples or high ex-
posure levels, the current study examined a relatively large sample size In conclusion, serum PFOA was found to be positively associated
with a general PFOA exposure level. In addition to the common con- with diabetes mellitus in men, independent of several covariates and
founders, we analysed other covariates that may have effects on the de- in accordance with emerging toxicology data. In particular, our findings
velopment of diabetes, such as PIR, education level, energy intake, time indicate that PFOA may disrupt cholesterol metabolism at environmen-
spent in front of a screen and serum cotinine. Furthermore, we carried tally relevant exposures. Considering the limitations of this study,

Fig. 3. Scatter plots and fitted lines with 95% confidence intervals (95% CI) for the relationship between PFOA concentrations and serum TC, for males aged 40–60 (A) and females aged
60–70 (B).
X. He et al. / Science of the Total Environment 625 (2018) 566–574 573

further prospective studies are needed to test the causality of this asso- Ferguson, K.K., O'Neill, M.S., Meeker, J.D., 2013. Environmental contaminant exposures
and preterm birth: a comprehensive review. J. Toxicol. Environ. Health B Crit. Rev.
ciation and elucidate the intermediate pathways involved. 16, 69–113.
Fisher, M., Arbuckle, T.E., Wade, M., Haines, D.A., 2013. Do perfluoroalkyl substances affect
metabolic function and plasma lipids?—analysis of the 2007–2009, Canadian Health
Acknowledgement Measures Survey (CHMS) Cycle 1. Environ. Res. 121, 95–103.
Fromme, H., Mosch, C., Morovitz, M., Alba-Alejandre, I., Boehmer, S., Kiranoglu, M., et al.,
This study was supported by grants from the National Natural Sci- 2010. Pre- and postnatal exposure to perfluorinated compounds (PFCs). Environ.
Sci. Technol. 44, 7123–7129.
ence Foundation of China (81370920, 81770773), Natural Science Foun-
Goldfield, G.S., Saunders, T.J., Kenny, G.P., Hadjiyannakis, S., Phillips, P., Alberga, A.S., et al.,
dation of Jiangsu Province (BK20171499), Project of "Six Talents Peak of 2013. Screen viewing and diabetes risk factors in overweight and obese adolescents.
Jiangsu Province (2013WSN-023)", Jiangsu Province's Key Medical Tal- Am. J. Prev. Med. 44, S364–370.
ents (co-construction) Program, Project of “333” Talent in Jiangsu Prov- Gore, A.C., Chappell, V.A., Fenton, S.E., Flaws, J.A., Nadal, A., Prins, G.S., et al., 2015. EDC-2:
the Endocrine Society's second scientific statement on endocrine-disrupting
ince, Jiangsu Province Official Hospital Scientific Research Initial chemicals. Endocr. Rev. 36, E1–E150.
Funding (RPF201501), and Jiangsu Province Official Hospital Talents Halban, P.A., Polonsky, K.S., Bowden, D.W., Hawkins, M.A., Ling, C., Mather, K.J., et al., 2014.
Construction Fund Research Project (IR2015101). β-Cell failure in type 2 diabetes: postulated mechanisms and prospects for preven-
tion and treatment. J. Clin. Endocrinol. Metab. 99, 1983–1992.
Hanssen, L., Dudarev, A.A., Huber, S., Odland, J.O., Nieboer, E., Sandanger, T.M., 2013. Par-
Conflicts of interest tition of perfluoroalkyl substances (PFASs) in whole blood and plasma, assessed in
maternal and umbilical cord samples from inhabitants of arctic Russia and
Uzbekistan. Sci. Total Environ. 447, 430–437.
The authors declare that there is no conflict of interest. He, K., Xun, P., Liu, K., Morris, S., Reis, J., Guallar, E., 2013. Mercury exposure in young
adulthood and incidence of diabetes later in life: the CARDIA Trace Element Study. Di-
Appendix A. Supplementary data abetes Care 36, 1584–1589.
Hemat, H., Wilhelm, M., Volkel, W., Mosch, C., Fromme, H., Wittsiepe, J., 2010. Low serum
levels of perfluorooctanoic acid (PFOA), perfluorooctane sulfonate (PFOS) and
Supplementary data to this article can be found online at https://doi. perfluorohexane sulfonate (PFHxS) in children and adults from Afghanistan. Sci.
org/10.1016/j.scitotenv.2017.12.186. Total Environ. 408, 3493–3495.
Hinderliter, P.M., Han, X., Kennedy, G.L., Butenhoff, J.L., 2006. Age effect on
perfluorooctanoate (PFOA) plasma concentration in post-weaning rats following
References oral gavage with ammonium perfluorooctanoate (APFO). Toxicology 225, 195–203.
Holst, C., Becker, U., Jorgensen, M.E., Gronbaek, M., Tolstrup, J.S., 2017. Alcohol drinking
Ahmadkhaniha, R., Mansouri, M., Yunesian, M., Omidfar, K., Jeddi, M.Z., Larijani, B., et al., patterns and risk of diabetes: a cohort study of 70,551 men and women from the gen-
2014. Association of urinary bisphenol a concentration with type-2 diabetes mellitus. eral Danish population. Diabetologia 60, 1941–1950.
J. Environ. Health Sci. Eng. 12, 64. Honda, T., Pun, V.C., Manjourides, J., Suh, H., 2017. Associations between long-term expo-
Alshaarawy, O., Zhu, M., Ducatman, A.M., Conway, B., Andrew, M.E., 2014. Urinary polycy- sure to air pollution, glycosylated hemoglobin and diabetes. Int. J. Hyg. Environ.
clic aromatic hydrocarbon biomarkers and diabetes mellitus. Occup. Environ. Med. Health 220, 1124–1132.
71, 437–441. IDF Diabetes Atlas Group, 2015. Update of mortality attributable to diabetes for the IDF
Arbuckle, T.E., Kubwabo, C., Walker, M., Davis, K., Lalonde, K., Kosarac, I., et al., 2013. Um- Diabetes Atlas: estimates for the year 2013. Diabetes Res. Clin. Pract. 109, 461–465.
bilical cord blood levels of perfluoroalkyl acids and polybrominated flame retardants. Jain, R.B., 2014. Contribution of diet and other factors to the levels of selected polyfluorinated
Int. J. Hyg. Environ. Health 216, 184–194. compounds: data from NHANES 2003–2008. Int. J. Hyg. Environ. Health 217, 52–61.
Audouze, K., Brunak, S., Grandjean, P., 2013. A computational approach to chemical etiol- James-Todd, T., Stahlhut, R., Meeker, J.D., Powell, S.G., Hauser, R., Huang, T., et al., 2012.
ogies of diabetes. Sci. Rep. 3, 2712. Urinary phthalate metabolite concentrations and diabetes among women in the Na-
Bainbridge, K.E., Hoffman, H.J., Cowie, C.C., 2008. Diabetes and hearing impairment in the tional Health and Nutrition Examination Survey (NHANES) 2001–2008. Environ.
United States: audiometric evidence from the National Health and Nutrition Exami- Health Perspect. 120, 1307–1313.
nation Survey, 1999 to 2004. Ann. Intern. Med. 149, 1–10. Jarvandi, S., Schootman, M., Racette, S.B., 2015. Breakfast intake among adults with type 2
Bao, W.W., Qian, Z.M., Geiger, S.D., Liu, E., Liu, Y., Wang, S.Q., et al., 2017. Gender-specific diabetes: influence on daily energy intake. Public Health Nutr. 18, 2146–2152.
associations between serum isomers of perfluoroalkyl substances and blood pressure Kang, J.S., Choi, J.S., Park, J.W., 2016. Transcriptional changes in steroidogenesis by
among Chinese: isomers of C8 Health Project in China. Sci. Total Environ. 607-608, perfluoroalkyl acids (PFOA and PFOS) regulate the synthesis of sex hormones in
1304–1312. H295R cells. Chemosphere 155, 436–443.
Bond, G.G., Dietrich, D.R., 2017. Further thoughts on limitations, uncertainties and com- Karnes, C., Winquist, A., Steenland, K., 2014. Incidence of type II diabetes in a cohort with
peting interpretations regarding chemical exposures and diabetes. J. Epidemiol. Com- substantial exposure to perfluorooctanoic acid. Environ. Res. 128, 78–83.
munity Health 71, 943. Kennedy Jr., G.L., Butenhoff, J.L., Olsen, G.W., O'Connor, J.C., Seacat, A.M., Perkins, R.G., et
Buck, R.C., Franklin, J., Berger, U., Conder, J.M., Cousins, I.T., de Voogt, P., et al., 2011. al., 2004. The toxicology of perfluorooctanoate. Crit. Rev. Toxicol. 34, 351–384.
Perfluoroalkyl and polyfluoroalkyl substances in the environment: terminology, clas- Kim, S.J., Heo, S.H., Lee, D.S., Hwang, I.G., Lee, Y.B., Cho, H.Y., 2016. Gender differences in
sification, and origins. Integr. Environ. Assess. Manag. 7, 513–541. pharmacokinetics and tissue distribution of 3 perfluoroalkyl and polyfluoroalkyl sub-
Buhrke, T., Kruger, E., Pevny, S., Rossler, M., Bitter, K., Lampen, A., 2015. Perfluorooctanoic stances in rats. Food Chem. Toxicol. 97, 243–255.
acid (PFOA) affects distinct molecular signalling pathways in human primary hepato- Kjeldsen, L.S., Bonefeld-Jorgensen, E.C., 2013. Perfluorinated compounds affect the func-
cytes. Toxicology 333, 53–62. tion of sex hormone receptors. Environ. Sci. Pollut. Res. Int. 20, 8031–8044.
Butt, C.M., Berger, U., Bossi, R., Tomy, G.T., 2010. Levels and trends of poly- and La Rocca, C., Tait, S., Guerranti, C., Busani, L., Ciardo, F., Bergamasco, B., et al., 2014. Exposure
perfluorinated compounds in the arctic environment. Sci. Total Environ. 408, to endocrine disrupters and nuclear receptor gene expression in infertile and fertile
2936–2965. women from different Italian areas. Int. J. Environ. Res. Public Health 11, 10146–10164.
Calafat, A.M., Wong, L.Y., Kuklenyik, Z., Reidy, J.A., Needham, L.L., 2007. Polyfluoroalkyl La Rocca, C., Tait, S., Guerranti, C., Busani, L., Ciardo, F., Bergamasco, B., et al., 2015. Expo-
chemicals in the U.S. population: data from the National Health and Nutrition Exam- sure to endocrine disruptors and nuclear receptors gene expression in infertile and
ination Survey (NHANES) 2003–2004 and comparisons with NHANES 1999–2000. fertile men from Italian areas with different environmental features. Int. J. Environ.
Environ. Health Perspect. 115, 1596–1602. Res. Public Health 12, 12426–12445.
Cho, C.R., Lam, N.H., Cho, B.M., Kannan, K., Cho, H.S., 2015. Concentration and correlations Lau, C., Anitole, K., Hodes, C., Lai, D., Pfahles-Hutchens, A., Seed, J., 2007. Perfluoroalkyl
of perfluoroalkyl substances in whole blood among subjects from three different geo- acids: a review of monitoring and toxicological findings. Toxicol. Sci. 99, 366–394.
graphical areas in Korea. Sci. Total Environ. 512-513, 397–405. Lebovitz, H.E., Banerji, M.A., 2001. Insulin resistance and its treatment by thiazolidinediones.
Christensen, K.Y., Raymond, M., Thompson, B.A., Anderson, H.A., 2016. Perfluoroalkyl sub- Recent Prog. Horm. Res. 56, 265–294.
stances in older male anglers in Wisconsin. Environ. Int. 91, 312–318. Leonard, R.C., Kreckmann, K.H., Sakr, C.J., Symons, J.M., 2008. Retrospective cohort mortal-
Cnop, M., Welsh, N., Jonas, J.C., Jorns, A., Lenzen, S., Eizirik, D.L., 2005. Mechanisms of pan- ity study of workers in a polymer production plant including a reference population
creatic beta-cell death in type 1 and type 2 diabetes: many differences, few similari- of regional workers. Ann. Epidemiol. 18, 15–22.
ties. Diabetes 54 (Suppl. 2), S97–107. Li, Y., Cheng, Y., Xie, Z., Zeng, F., 2017. Perfluorinated alkyl substances in serum of the
Conway, B., Innes, K.E., Long, D., 2016. Perfluoroalkyl substances and beta cell deficient di- southern Chinese general population and potential impact on thyroid hormones.
abetes. J. Diabetes Complicat. 30, 993–998. Sci. Rep. 7, 43380.
Costa, G., Sartori, S., Consonni, D., 2009. Thirty years of medical surveillance in Lin, C.Y., Chen, P.C., Lin, Y.C., Lin, L.Y., 2009. Association among serum perfluoroalkyl
perfluooctanoic acid production workers. J. Occup. Environ. Med. 51, 364–372. chemicals, glucose homeostasis, and metabolic syndrome in adolescents and adults.
Domazet, S.L., Grontved, A., Timmermann, A.G., Nielsen, F., Jensen, T.K., 2016. Longitudinal Diabetes Care 32, 702–707.
associations of exposure to perfluoroalkylated substances in childhood and adoles- Lind, L., Zethelius, B., Salihovic, S., van Bavel, B., Lind, P.M., 2014. Circulating levels of
cence and indicators of adiposity and glucose metabolism 6 and 12 years later: the perfluoroalkyl substances and prevalent diabetes in the elderly. Diabetologia 57,
European Youth Heart Study. Diabetes Care 39, 1745–1751. 473–479.
Eriksen, K.T., Raaschou-Nielsen, O., McLaughlin, J.K., Lipworth, L., Tjonneland, A., Overvad, Liu, H.S., Wen, L.L., Chu, P.L., Lin, C.Y., 2017. Association among total serum isomers of
K., et al., 2013. Association between plasma PFOA and PFOS levels and total cholester- perfluorinated chemicals, glucose homeostasis, lipid profiles, serum protein and met-
ol in a middle-aged Danish population. PLoS One 8, e56969. abolic syndrome in adults: NHANES, 2013–2014. Environ. Pollut. 232, 73–79.
574 X. He et al. / Science of the Total Environment 625 (2018) 566–574

Lorenzo, C., Williams, K., Hunt, K.J., Haffner, S.M., 2007. The National Cholesterol Educa- Saldana, T.M., Basso, O., Hoppin, J.A., Baird, D.D., Knott, C., Blair, A., et al., 2007. Pesticide
tion Program - Adult Treatment Panel III, International Diabetes Federation, and exposure and self-reported gestational diabetes mellitus in the Agricultural Health
World Health Organization definitions of the metabolic syndrome as predictors of in- Study. Diabetes Care 30, 529–534.
cident cardiovascular disease and diabetes. Diabetes Care 30, 8–13. Steenland, K., Woskie, S., 2012. Cohort mortality study of workers exposed to
Lupi, R., Del Guerra, S., Marselli, L., Bugliani, M., Boggi, U., Mosca, F., et al., 2004. perfluorooctanoic acid. Am. J. Epidemiol. 176, 909–917.
Rosiglitazone prevents the impairment of human islet function induced by fatty Steenland, K., Tinker, S., Frisbee, S., Ducatman, A., Vaccarino, V., 2009. Association of
acids: evidence for a role of PPARgamma2 in the modulation of insulin secretion. perfluorooctanic acid (PFOA) and perfluoroctanesulfonate (PFOS) with serum lipids
Am. J. Physiol. Endocrinol. Metab. 286, E560–567. among adults living near a chemical plant. 20.
MacNeil, J., Steenland, N.K., Shankar, A., Ducatman, A., 2009. A cross-sectional analysis of Su, T.C., Kuo, C.C., Hwang, J.J., Lien, G.W., Chen, M.F., Chen, P.C., 2016. Serum
type II diabetes in a community with exposure to perfluorooctanoic acid (PFOA). En- perfluorinated chemicals, glucose homeostasis and the risk of diabetes in working-
viron. Res. 109, 997–1003. aged Taiwanese adults. Environ. Int. 88, 15–22.
Mamtani, M., Curran, J.E., Blangero, J., Kulkarni, H., 2016. Association of Urinary phthalates Suh, K.S., Choi, E.M., Kim, Y.J., Hong, S.M., Park, S.Y., Rhee, S.Y., et al., 2017.
with self-reported eye affliction/retinopathy in individuals with diabetes: National Perfluorooctanoic acid induces oxidative damage and mitochondrial dysfunction in
Health and Nutrition Examination Survey, 2001–2010. J. Diabetes Res. 2016, pancreatic beta-cells. Mol. Med. Rep. 15, 3871–3878.
7269896. Thiering, E., Bruske, I., Kratzsch, J., Thiery, J., Sausenthaler, S., Meisinger, C., et al., 2011.
Menke, A., Orchard, T.J., Imperatore, G., Bullard, K.M., Mayer-Davis, E., Cowie, C.C., 2013. Prenatal and postnatal tobacco smoke exposure and development of insulin resis-
The prevalence of type 1 diabetes in the United States. Epidemiology 24, 773–774. tance in 10 year old children. Int. J. Hyg. Environ. Health 214, 361–368.
Moon, S.S., 2014. Additive effect of heavy metals on metabolic syndrome in the Korean Toms, L.M., Calafat, A.M., Kato, K., Thompson, J., Harden, F., Hobson, P., et al., 2009.
population: the Korea National Health and Nutrition Examination Survey Polyfluoroalkyl chemicals in pooled blood serum from infants, children, and adults
(KNHANES) 2009–2010. Endocrine 46, 263–271. in Australia. Environ. Sci. Technol. 43, 4194–4199.
Moyer, J., Wilson, D., Finkelshtein, I., Wong, B., Potts, R., 2012. Correlation between sweat Wang, Z., Cousins, I.T., Scheringer, M., Hungerbuhler, K., 2013. Fluorinated alternatives to
glucose and blood glucose in subjects with diabetes. Diabetes Technol. Ther. 14, long-chain perfluoroalkyl carboxylic acids (PFCAs), perfluoroalkane sulfonic acids
398–402. (PFSAs) and their potential precursors. Environ. Int. 60, 242–248.
Nelson, J.W., Hatch, E.E., Webster, T.F., 2010. Exposure to polyfluoroalkyl chemicals and Wang, X., Liu, L., Zhang, W., Zhang, J., Du, X., Huang, Q., et al., 2017. Serum metabolome
cholesterol, body weight, and insulin resistance in the general U.S. population. Envi- biomarkers associate low-level environmental perfluorinated compound exposure
ron. Health Perspect. 118, 197–202. with oxidative/nitrosative stress in humans. Environ. Pollut. 229, 168–176.
Ng, C.A., Hungerbuehler, K., 2015. Exploring the use of molecular docking to identify Wolf, C.J., Takacs, M.L., Schmid, J.E., Lau, C., Abbott, B.D., 2008. Activation of mouse and
bioaccumulative perfluorinated alkyl acids (PFAAs). Environ. Sci. Technol. 49, human peroxisome proliferator-activated receptor alpha by perfluoroalkyl acids of
12306–12314. different functional groups and chain lengths. Toxicol. Sci. 106, 162–171.
Olsen, G.W., Burris, J.M., Ehresman, D.J., Froehlich, J.W., Seacat, A.M., Butenhoff, J.L., et al., 2007. Xu, C., Liu, Q., Zhang, Q., Gu, A., Jiang, Z.Y., 2015. Urinary enterolactone is associated with
Half-life of serum elimination of perfluorooctanesulfonate,perfluorohexanesulfonate, obesity and metabolic alteration in men in the US National Health and Nutrition Ex-
and perfluorooctanoate in retired fluorochemical production workers. Environ. Health amination Survey 2001–10. Br. J. Nutr. 113, 683–690.
Perspect. 115, 1298–1305. Yan, S., Zhang, H., Zheng, F., Sheng, N., Guo, X., Dai, J., 2015. Perfluorooctanoic acid expo-
Peng, Q., Harlow, S.D., Park, S.K., 2015. Urinary arsenic and insulin resistance in US adoles- sure for 28 days affects glucose homeostasis and induces insulin hypersensitivity in
cents. Int. J. Hyg. Environ. Health 218, 407–413. mice. Sci. Rep. 5, 11029.
Poet, T.S., Schlosser, P.M., Rodriguez, C.E., Parod, R.J., Rodwell, D.E., Kirman, C.R., 2016. Zhang, Y., Jiang, W., Fang, S., Zhu, L., Deng, J., 2014. Perfluoroalkyl acids and the isomers of
Using physiologically based pharmacokinetic modeling and benchmark dose perfluorooctanesulfonate and perfluorooctanoate in the sera of 50 new couples in
methods to derive an occupational exposure limit for N-methylpyrrolidone. Regul. Tianjin, China. Environ. Int. 68, 185–191.
Toxicol. Pharmacol. 76, 102–112. Zhang, Z., Kris-Etherton, P.M., Hartman, T.J., 2014. Birth weight and risk factors for cardio-
Post, G.B., Cohn, P.D., Cooper, K.R., 2012. Perfluorooctanoic acid (PFOA), an emerging vascular disease and type 2 diabetes in US children and adolescents: 10 year results
drinking water contaminant: a critical review of recent literature. Environ. Res. 116, from NHANES. Matern. Child Health J. 18, 1423–1432.
93–117. Zhang, C., Sundaram, R., Maisog, J., Calafat, A.M., Barr, D.B., Buck Louis, G.M., 2015. A pro-
Rhee, S.Y., Hwang, Y.C., Woo, J.T., Chin, S.O., Chon, S., Kim, Y.S., 2013. Arsenic exposure and spective study of prepregnancy serum concentrations of perfluorochemicals and the
prevalence of diabetes mellitus in Korean adults. J. Korean Med. Sci. 28, 861–868. risk of gestational diabetes. Fertil. Steril. 103, 184–189.
Sakr, C.J., Kreckmann, K.H., Green, J.W., Gillies, P.J., Reynolds, J.L., Leonard, R.C., 2007.
Cross-sectional study of lipids and liver enzymes related to a serum biomarker of ex-
posure (ammonium perfluorooctanoate or APFO) as part of a general health survey in
a cohort of occupationally exposed workers. J. Occup. Environ. Med. 49, 1086–1096.

You might also like