Skip main navigation

Per- and Polyfluoroalkyl Substances and Incident Hypertension in Multi-Racial/Ethnic Women: The Study of Women's Health Across the Nation

Originally publishedhttps://doi.org/10.1161/HYPERTENSIONAHA.121.18809Hypertension. 2022;79:1876–1886

Abstract

Background:

Perfluoroalkyl and polyfluoroalkyl substances (PFAS) are ubiquitous synthetic chemicals that may disrupt blood pressure controls; however, human evidence to support this hypothesis is scant. We examined the association between serum concentrations of PFAS and risks of developing hypertension.

Methods:

This study included 1058 midlife women initially free of hypertension from the multiracial and multiethnic SWAN (Study of Women’s Health Across the Nation) with annual follow-up visits between 1999 and 2017. Hypertension was defined as blood pressure ≥140 mm Hg systolic or ≥90 mm Hg diastolic or receiving antihypertensive treatment. Cox proportional hazards models were utilized to calculate hazard ratios and 95% CIs. Quantile g-computation was implemented to evaluate the joint effect of PFAS mixtures.

Results:

During 11 722 person-years of follow-up, 470 participants developed incident hypertension (40.1 cases per 1000 person-years). Compared with the lowest tertile, women in the highest tertile of baseline serum concentrations had adjusted hazard ratios of 1.42 (95% CI, 1.19–1.68) for perfluorooctane sulfonate (P trend=0.01), 1.47 (95% CI, 1.24–1.75) for linear perfluorooctanoate (P trend=0.01), and 1.42 (95% CI, 1.19–1.70) for 2-(N-ethyl-perfluorooctane sulfonamido) acetate (P trend=0.01). No significant associations were observed for perfluorononanoate and perfluorohexane sulfonate. In the mixture analysis, women in the highest tertile of overall PFAS concentrations had a hazard ratio of 1.71 (95% CI, 1.15–2.54; P trend=0.008), compared with those in the lowest tertile.

Conclusions:

Several PFAS showed positive associations with incident hypertension. These findings suggest that PFAS might be an underappreciated contributing factor to women’s cardiovascular disease risk.

Novelty and Relevance

What Is New?

  • This is the first study to evaluate the effects of perfluoroalkyl and polyfluoroalkyl substances on incident hypertension among midlife women, using a prospective cohort of 1,058 women initially free of hypertension with annual follow-up visits from 1999 to 2017.

What Is Relevant?

  • Higher perfluorooctane sulfonate, perfluorooctanoate, and 2-(N-ethyl-perfluorooctane sulfonamido) acetate are associated with 42%, 47%, and 42% higher risks of hypertension, respectively, comparing the highest to the lowest tertile.

  • Participants with higher overall perfluoroalkyl and polyfluoroalkyl substances concentrations are associated with a 71% increase in the risk of hypertension.

Clinical/Pathophysiological Implications?

Perfluoroalkyl and polyfluoroalkyl substances are potentially modifiable risk factors for hypertension and cardiovascular disease. Our findings are of significant public health concern since perfluoroalkyl and polyfluoroalkyl substances are ubiquitous in humans and the environment.

Human exposure to per- and polyfluoroalkyl substance (PFAS) is of significant public health concern because PFAS are persistent in the environment and many also persist in humans.1 PFAS, synthetic chemicals widely used in industrial applications and consumer products, have been estimated to contaminate drinking water consumed by more than 200 million Americans.2 Data from the National Health and Nutrition Examination Survey demonstrate the ubiquity of PFAS exposure, as nearly all Americans have detectable concentrations of at least one PFAS in their blood.3 Due to their structural similarity to fatty acids, PFAS may interfere with the action of PPARs (peroxisome proliferator-activated receptors), specifically PPAR-α and PPAR-γ, and their signaling pathways, and thus, modulate metabolic pathways.4,5 Toxicological studies have also revealed that PFAS exposure is related to oxidative stress and endothelial dysfunction.6,7 As a result, the cardiovascular system is vulnerable to PFAS toxicity. PFAS have been associated with higher risks of cardiovascular disease (CVD) and peripheral arterial disease,8,9 as well as several CVD risk factors including thyroid disease,10–12 elevated total cholesterol and low-density lipoprotein levels,13–15 higher body mass index,16–20 and altered glucose homeostasis.21–23 However, far less is known about whether PFAS may disrupt blood pressure controls and increase the risk of developing hypertension.

Hypertension, a condition that affects >1 billion adults worldwide, is one of the most important risk factors for CVD.24 Hypertension is also the leading cause of death and disability-adjusted life-years globally.25 Until recently, only one study has investigated the associations between PFAS exposure and incident hypertension,26 with no relationship detected in a prospective cohort of 660 prediabetic and overweight adults. Three cross-sectional studies have linked certain PFAS to increased prevalence of hypertension and elevated blood pressure,27–29 but 3 separate studies reported no association or even suggested protective effects of PFAS.30–32

Given the ubiquitous nature of PFAS exposure, even a modest association between PFAS and hypertension would place a large number of people at increased risk for cardiovascular morbidity and mortality. To better understand the relationship between PFAS and hypertension, we used data from the SWAN (Study of Women’s Health Across the Nation), which is a prospective study with approximately annual follow-up visits over 18 years. The menopausal transition marks a period of decline in follicle numbers and characteristic fluctuations in hormone levels. Furthermore, the menopausal transition is well-described as a period of increasing hypertension and cardiovascular disease risk.33 The present study examined PFAS individually, and jointly as mixtures. We hypothesized that higher serum PFAS concentrations would be associated with higher risks of incident hypertension.

Methods

Study Design and Study Population

SWAN is a multisite, multiracial, and multiethnic, community-based longitudinal study designed to characterize physiological and psychosocial changes among women transitioning through menopause. The study design and procedures, including recruitment and enrollment, have been described in detail elsewhere.34 Between November 1995 and October 1997, a total of 3302 premenopausal or early perimenopausal women aged 42 to 52 years were recruited from seven geographically distinct sites across the United States. All sites enrolled Non-Hispanic White women in addition to one additional racial and ethnic group. For the Boston, MA; Chicago, IL; Pittsburgh, PA; and Southeast Michigan, Black women were enrolled, whereas Chinese women were enrolled at the Oakland, CA, Hispanic women from Newark, NJ; Chicago, IL, and Japanese women from Los Angeles, CA. Eligibility criteria for SWAN included age 42 to 52 years upon entry, presence of an intact uterus, at least one menstrual period in the prior 3 months, and not taking hormone medications within the prior 3 months. All participants provided written informed consent at each study visit, and all protocols used in SWAN were approved by the institutional review board at each participating site.

The SWAN MPS (Multi-Pollutant Study) was initiated in 2016 to examine metabolic and reproductive health effects of multiple environmental pollutants including PFAS in midlife women. The study designs of the SWAN MPS were described elsewhere.35–37 We used repository samples available from the third SWAN follow-up (visit 03 or the MPS baseline, 1999–2000) for environmental exposure assessments (N=2694). MPS required women to have both blood and urine samples. Hispanic women were not eligible because urine samples were not collected (n=646). We also excluded women with insufficient serum or urine samples (n=648). Among 1400 SWAN MPS participants, we excluded women with missing information on hypertension (n=18), those who had already had hypertension (n=306) at MPS baseline (visit 03), and those with missing values for other covariates (n=18), yielding a final analytic sample of 1058 followed up from 1999 to 2017 (Figure S1).

Participant characteristics were significantly different between those included versus those not included in the SWAN MPS due to insufficient biospecimens (Table S1). Therefore, we used inverse probability weighting to address the issue of potential selection bias. The details of inverse probability weighting are shown in the Supplemental Material.

Outcomes

Information on blood pressure and antihypertensive medication use was collected at each visit (approximately annually). The present study included data from 1999 to 2000 (SWAN follow-up visit 3, MPS baseline) through 2016 to 2017 (SWAN follow-up visit 16), with up to 13 time points included in the analysis. Blood pressure was measured according to a standardized protocol, with readings taken on the right arm, with the respondent seated and feet flat on the floor for at least 5 minutes before measurement. Respondents were asked not to smoke or consume any caffeinated beverage within 30 minutes of blood pressure measurement. Appropriate cuff size was determined based on arm circumference. A standard mercury sphygmomanometer was used to record systolic blood pressure (SBP) and diastolic blood pressure (DBP) at the first and fifth phase Korotkoff sounds. Two sequential blood pressure values were completed, with a minimum 2-minute rest period between measures. The average of these measures was employed in these analyses. Hypertension was defined as SBP≥140 mm Hg or DBP≥90 mm Hg or use of antihypertensive medications, in accordance with the definitions used in the sixth and seventh reports of the Joint National Committee on Prevention, Detection, Treatment and Control of High Blood Pressure.38,39

Assessment of Serum PFAS Concentrations

Serum samples collected at the MPS baseline (1999–2000) were sent to the Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention. The Centers for Disease Control and Prevention laboratory’s involvement did not constitute engagement in human-subjects research. We quantified perfluorohexane sulfonate (PFHxS), linear perfluorooctane sulfonate (n-PFOS), sum of branched isomers of PFOS (Sm-PFOS), linear perfluorooctanoate (n-PFOA), sum of branched PFOA, perfluorononanoate, perfluorodecanoate, perfluoroundecanoate, perfluorododecanoate, 2-(N-ethyl-perfluorooctane sulfonamido) acetate (EtFOSAA), and 2-(N-methyl-perfluorooctane sulfonamido) acetate (MeFOSAA) in 0.1 mL of serum, using an online solid-phase extraction-high performance liquid chromatography-isotope dilution-tandem mass spectrometry.40 The analytic methods and quality control procedures have been described elsewhere.36 The coefficient of variation of low- and high-quality controls ranged from 6% to 12%, depending on the analyte. The limit of detection was 0.1 ng/mL for all the analytes. Concentrations below the limit of detection were substituted with limit of detection.41 Total PFOS (PFOS) was computed as the sum of n-PFOS and Sm-PFOS.

Covariates

Given careful review of the literature for important variables associated with both PFOS exposure and hypertension risk,35,42 we selected a comprehensive set of confounders. We obtained the following data from participants’ standardized interviews conducted at each visit. Sociodemographic variables included age at baseline (in years), self-defined race and ethnicity (Black, Chinese, Japanese, White), study site (Southeast Michigan, Boston, Pittsburgh, Oakland, Los Angeles), education (high school or less, some college, college degree or higher), and financial strain (very hard, somewhat hard, not hard at all). Time-varying covariates included self-reported lifestyle factors, namely smoking status (never smoker, past smoker, current smoker), environmental tobacco smoking (0, 1–4, ≥5 person-hours),43 total calorie intake (in kcal), and alcohol intake (<1 drink/mo, >1 drink/mo and ≤1/wk, >1 drink/wk). Physical activity was assessed in various domains, including sports/exercise, household/caregiving, and daily routine, with a range of 3 to 15 (15 indicating the highest level of activity).44 Menopausal status was categorized into surgical postmenopausal, natural postmenopausal, late perimenopausal, early perimenopausal, premenopausal, or unknown due to hormone therapy using bleeding patterns and information about hormone therapy use. The SWAN has a small proportion (<5%) of missing data on time-varying covariates. To deal with missing data, we simply carried forward last observations.

Statistical Analysis

Descriptive statistics were calculated for participant characteristics and serum concentrations of PFAS at MPS baseline. Spearman correlation coefficients were calculated among multiple PFAS compounds. Cox proportional hazards regression model were utilized to calculate hazard ratio (HR) and 95% CI. We employed age instead of follow-up time as the time scale.45 Serum concentrations of PFAS were categorized into tertiles. HRs and 95% CIs were calculated by comparing the second and the highest tertiles of PFAS concentrations with the lowest tertile (the reference). We also examined the log-linear relationships using log2-transformed PFAS concentrations. In this case, HRs and 95% CIs were interpreted as changes per doubling in PFAS concentrations. Covariates considered in multivariable adjustments included race and ethnicity, educational attainment, financial strain, smoking status, environmental tobacco smoke, alcohol consumption, total calorie intake, and menopausal status. We included in data analyses n-PFOS, Sm-PFOS, n-PFOA, PFHxS, perfluorononanoate, EtFOSAA, and MeFOSAA that were detected in >97% of samples. Sum of branched PFOA, perfluorodecanoate, and perfluoroundecanoate with relatively low detection frequencies (<50%) were modeled as detect versus nondetect. perfluorododecanoate with extremely low detection frequencies (3.7%) was not included in the analysis. As the menopausal transition contributes to the increase in hypertension risks,33 we also examined the whether the relationships between PFAS and hypertension differed by menopausal status, by including interaction terms between PFAS and menopausal status (premenopause, perimenopause, and natural postmenopause).

As an additional exploratory analysis, we utilized quantile g-computation to evaluate the overall effects of exposure to PFAS mixtures on incident hypertension.46 Quantile g-computation is a valuable tool designed for estimating the joint effects of correlated chemical mixtures. This approach combined the inferential ability of weighted quantile sum regression47 with the flexibility of g-computation, a method of causal effect estimation. In the current study, all PFAS mixture components were first transformed into tertiles. Then a Cox proportional hazards regression was fit for PFAS, covariates, and incident hypertension, which defined the weights for each PFAS. Weights represented the association between each PFAS and incident hypertension. The sum of weights was constructed as the quantile g-computation estimator. The mixture effect of PFAS was calculated using the quantile g-computation estimator and interpreted as the HR and 95% CI per tertile increase in all PFAS concentrations, adjusting for covariates described above for the individual PFAS models. To be consistent with the individual associations, we calculated and interpreted HRs comparing the highest and second tertiles to the lowest tertile. The mixture analyses were conducted in the R package qgcomp. Given that the qgcomp.cox function in the qgcomp package may yield invalid results using counting-process style data, we decided to use wide-format data to conduct quantile g-computation as an exploratory analysis. Details of the quantile g-computation and R codes are provided in the Supplemental Material.

Additionally, we performed a sensitivity analysis with further adjustment for body mass index and weight change over time. We did not consider body mass index and weight change over time as a confounder in the primary analysis in case of over-adjustment bias because PFAS may contribute to increased body mass index and weight gain.16–20,48 Furthermore, we evaluated the association between PFAS and changes in SBP and DBP in women without antihypertensive medication use or hypertension using linear mixed models to understand which of 2 measurements might drive the development of hypertension. Moreover, we modeled time to hypertension using parametric method, that is, accelerated failure time models, to examine the robustness of our findings. Finally, we replicated the analysis using wide-format data with baseline covariates rather than counting-process data. All the analyses were performed using SAS, version 9.4 (SAS Institute, Inc, Cary, NC) and R (version 4.0.0. R Foundation for Statistical Computing).

Results

At MPS baseline, the median age was 49.2 years, 54.5% were White women, 16.8% held a high school diploma or had not completed high school, 29.9% reported being very hard or somewhat hard paying for basics, 35.7% were past or current smokers, 37.8% were exposed to environmental tobacco smoke, and 24.9% were high-frequency alcohol drinkers (Table 1). In addition, 30% of women were postmenopausal. The median serum concentrations of PFAS at MPS baseline were 24.1 (interquartile range [IQR], 17.3–34.8) ng/mL for PFOS, 17.2 (IQR, 12.2–24.1) ng/mL for n-PFOS, 7.1 (IQR, 4.6–10.7) ng/mL for Sm-PFOS, 4.1 (IQR, 2.8–5.9) ng/mL for n-PFOA, 1.5 (IQR, 1.0–2.4) ng/mL for PFHxS, 0.6 (IQR, 0.4–0.8) ng/mL for perfluorononanoate, 1.2 (IQR, 0.6–2.2) ng/mL for EtFOSAA, and 1.4 (IQR, 0.9–2.3) ng/mL for MeFOSAA (Table S2). Spearman correlation coefficients among these compounds ranged from 0.11 to 0.82 (Figure S2). Of the 1058 participants, 470 women developed hypertension over 11 722 person-years (40.1 cases per 1000 person-years). The median follow-up time was 12.4 (IQR, 5.1–16.4) years.

Table 1. Baseline Characteristics and Blood Pressure in Midlife Women From the Study of Women’s Health Across the Nation Multi-Pollutant Study (n=1058)

CharacteristicsMedian (IQR) or N (%)
Age, y49.2 (47.2, 51.3)
Race and ethnicity
  White577 (54.5%)
 Black161 (15.2%)
 Chinese149 (14.1%)
 Japanese171 (16.2%)
Study site
 Southeast Michigan149 (14.1%)
 Boston172 (16.2%)
 Oakland261 (24.7%)
 Los Angeles299 (28.3%)
 Pittsburgh177 (16.7%)
Education
 ≤High school177 (16.7%)
 High school317 (30.0%)
 College273 (25.8%)
 Postcollege291 (27.5%)
Difficulty paying for basics
 Very hard58 (5.5%)
 Somewhat hard257 (24.3%)
 Not hard at all753 (70.2%)
Smoking status
 Never smoked681 (64.4 %)
 Past smoker278 (26.3%)
 Current smoker99 (9.4%)
Environmental tobacco smoke
 0 person-hours658 (62.2%)
 1–4 person-hours238 (22.5%)
 ≥5 person-hours162 (15.3%)
Alcohol intake
 None/low use538 (50.8%)
 Moderate use257 (24.3%)
 High use263 (24.9%)
Physical activity score8.0 (6.7–9.1)
Menopausal status
 Surgical post30 (2.8%)
 Natural post114 (10.8%)
 Late perimenopausal78 (7.4%)
 Early perimenopausal548 (51.8%)
 Premenopausal138 (13.0%)
 Unknown with hormone therapy150 (14.2%)
Total calorie intake, kcal1694 (1352–2161)
BMI, kg/m225.1 (22.1–29.9)
Systolic blood pressure, mm Hg108 (101–117)
Diastolic blood pressure, mm Hg71 (66–77)

BMI indicates body mass index; and IQR, interquartile range.

We found positive associations of PFOS, n-PFOA, and EtFOSAA with hypertension incidence (Table 2). Comparing the highest tertile of serum concentrations with the lowest, HRs of hypertension were 1.42 (95% CI, 1.19–1.68) for PFOS (P for trend<0.0001), 1.40 (95% CI, 1.18–1.66) for n-PFOS (P for trend<0.0001), 1.49 (95% CI, 1.25–1.78) for Sm-PFOS (P for trend<0.0001), 1.47 (95% CI, 1.24–1.75) for n-PFOA (P for trend<0.0001), and 1.42 (95% CI, 1.19–1.70) for EtFOSAA (P for trend=0.0003), after adjusting for race and ethnicity, study site, education, financial strain, smoking status, environmental tobacco smoke, alcohol consumption, total calorie intake, and menopausal status. Adjusted survival curves for each PFAS are shown in Figures S3 through S10. We also detected significant log-linear relationships of PFOS, EtFOSAA, and MeFOSAA with incident hypertension. Multivariable-adjusted HRs for a doubling in serum concentrations were 1.18 (95% CI, 1.09–1.28) for total PFOS, 1.17 (95% CI, 1.08–1.27) for n-PFOS, 1.16 (95% CI, 1.08–1.25) for Sm-PFOS, 1.17 (95% CI, 1.07–1.27) for n-PFOA, 1.09 (95% CI, 1.04–1.15) for EtFOSAA, and 1.10 (95% CI, 1.03–1.19) for MeFOSAA. Although we found significant associations of PFOS, n-PFOA, and EtFOSAA with hypertension, we observed no interactions of these chemicals with menopausal status (Table S3), except for MeFOSAA. The HR of hypertension was 1.36 (95% CI, 1.12–1.65) for premenopause, 0.98 (95% CI, 0.89–1.08) for perimenopause, and 1.34 (95% CI, 1.08–1.67) for natural postmenopause (P for interaction=0.0009).

Table 2. HR (95% CI) of Incident Hypertension With Tertile Changes and Per Doubling in Serum Concentrations of PFAS

PFASTertile of PFAS concentrationsP value for trendPer doubling increase HR (95% CI)P value
Tertile 1 HR (95% CI)Tertile 2 HR (95% CI)Tertile 3 HR (95% CI)
PFOS
 Median (IQR), ng/mL14.5 (11.6–17.1)24.0 (21.8–27.2)41.9 (34.8–53.9)
 No. cases/person-years133/4212156/4009181/3501
 Model 1*Ref1.23 (1.03–1.46)1.54 (1.31–1.82)<0.00011.21 (1.13–1.31)<0.0001
 Model 2Ref1.11 (0.93–1.33)1.42 (1.19–1.68)<0.00011.18 (1.09–1.28)<0.0001
n-PFOS
 Median (IQR), ng/mL10.5 (8.3–12.2)17.2 (15.7–19.0)28.6 (24.2–37.6)
 No. of cases/person-years133/4096152/4166185/3460
 Model 1*Ref1.14 (0.96–1.35)1.50 (1.27–1.77)<0.00011.20 (1.11–1.30)<0.0001
 Model 2Ref1.00 (0.84–1.19)1.40 (1.18–1.66)<0.00011.17 (1.08–1.27)0.0002
Sm-PFOS
 Median (IQR), ng/mL3.9 (2.9–4.6)7.1 (6.2–8.1)13.1 (10.7–17.2)
 No. cases/person-years133/4202156/3867181/3653
 Model 1*Ref1.24 (1.05–1.48)1.63 (1.37–1.92)<0.00011.19 (1.11–1.28)<0.0001
 Model 2Ref1.14 (0.95–1.36)1.49 (1.25–1.78)<0.00011.16 (1.08–1.25)<0.0001
n-PFOA
 Median (IQR), ng/mL2.4 (1.9–2.9)4.1 (3.7–4.5)6.8 (5.8–8.9)
 No. cases/person-years141/4103150/3900179/3719
 Model 1*Ref1.34 (1.13–1.59)1.52 (1.29–1.80)<0.00011.19 (1.10–1.29)<0.0001
 Model 2Ref1.37 (1.15–1.63)1.47 (1.24–1.75)<0.00011.17 (1.07–1.27)0.0005
PFNA
 Median (IQR), ng/mL0.3 (0.3–0.4)0.6 (0.5–0.7)1.0 (0.9–1.1)
 No. cases/person-years138/3708188/4796144/3218
 Model 1*Ref0.95 (0.81–1.11)1.05 (0.89–1.25)0.531.06 (0.98–1.15)0.17
 Model 2Ref0.93 (0.79–1.09)1.00 (0.83–1.19)0.971.05 (0.97–1.14)0.24
PFHxS
 Median (IQR), ng/mL0.8 (0.6–1.0)1.5 (1.3–1.7)3.2 (2.4–4.8)
 No. cases/person-years150/3854149/4005171/3863
 Model 1*Ref0.86 (0.72–1.02)1.06 (0.90–1.25)0.370.99 (0.94–1.04)0.71
 Model 2Ref0.84 (0.71–1.00)1.06 (0.89–1.25)0.380.98 (0.93–1.04)0.56
EtFOSAA
 Median (IQR), ng/mL0.5 (0.4–0.6)1.2 (0.9–1.4)3.0 (2.3–4.7)
 No. cases/person-years127/4136164/3996179/3590
 Model 1*Ref1.47 (1.24–1.76)1.57 (1.32–1.87)<0.00011.12 (1.07–1.18)<0.0001
 Model 2Ref1.39 (1.16–1.66)1.42 (1.19–1.70)0.00031.09 (1.04–1.15)0.0005
MeFOSAA
 Median (IQR), ng/mL0.7 (0.5–0.9)1.4 (1.2–1.7)2.8 (2.3–3.6)
 No. cases/person-years134/4016149/4027187/3679
 Model 1*Ref0.96 (0.81–1.15)1.24 (1.04–1.48)0.0061.14 (1.06–1.22)0.0004
 Model 2Ref0.91 (0.76–1.09)1.15 (0.96–1.37)0.061.10 (1.03–1.19)0.009

HRs (95% CIs) were computed from Cox proportional hazards model with age as the time scale. EtFOSAA indicates 2-(N-ethyl-perfluorooctane sulfonamido) acetate; HR, hazard ratio; IQR, interquartile range; MeFOSAA, 2-(N-methyl-perfluorooctane sulfonamido) acetate; n-PFOA, linear perfluorooctanoate; n-PFOS, linear perfluorooctane sulfonate; PFAS, perfluoroalkyl and polyfluoroalkyl substances; PFHxS, perfluorohexane sulfonate; PFOA, perfluorooctanoate; PFOS, perfluorooctane sulfonate; Ref, reference; and Sm-PFOS, sum of branched isomers of perfluorooctane sulfonate.

* Model 1 was adjusted for race and ethnicity and study site with inverse probability weight.

† Model 2: Model 1+education, financial strain, smoking status, environmental tobacco smoke, alcohol consumption, total calorie intake, and menopausal status with inverse probability weight.

In the sensitivity analyses, the effect estimates remained similar after further adjustment for weight change over time (Tables S4 and S5). No significant associations were observed for sum of branched PFOA, perfluorodecanoate, and perfluoroundecanoate with relatively low detection frequencies (Table S6). Furthermore, when examining blood pressure among women before receiving antihypertensive medications or developing hypertension, we found significant associations of PFOS, EtFOSAA, and MeFOSAA with SBP and DBP (Tables S7 and S8). Using parametric accelerated failure time models, we observed significant associations of PFOS, n-PFOS, Sm-PFOS, n-PFOA, and EtFOSAA with shorter time to the development of hypertension (Table S9).

The quantile g-computation estimated beta coefficients for all PFAS, as shown in Table S6, including n-PFOS (β=0.051), Sm-PFOS (β=0.115), EtFOSAA (β=0.049), MeFOSAA (β=0.0092), n-PFOA (β=0.032), perfluorononanoate (β=0.086), and PFHxS (β=−0.074). These beta coefficients were then incorporated as the weights into the construction of quantile g-computation estimator. Compared with those with the lowest tertile of overall PFAS concentrations, women with the second tertile and highest tertile were 1.31 (95% CI, 1.07–1.59) and 1.71 (95% CI, 1.15–2.54) times more likely to develop hypertension during the follow-up (P for trend=0.008), respectively (Figure 1).

Figure.

Figure. Adjusted survival curves for incident hypertension by tertiles of serum concentrations of perfluoroalkyl and polyfluoroalkyl substances mixtures from quantile g-computation. The model was adjusted for race and ethnicity, study site, education, financial strain, smoking status, environmental tobacco smoke, alcohol consumption, total calorie intake, and menopausal status, with inverse probability weighting. The adjusted hazard ratio of incident hypertension was 1.31 (95% CI, 1.07–1.59) for tertile 2 and 1.71 (95% CI, 1.15–2.54) for tertile 3, compared with those in tertile 1 (P for trend=0.008).

Discussion

In this 18-year prospective cohort of 1058 women with 11 722 person-years of observations in annual follow-up visits, we found that PFOS, n-PFOA, EtFOSAA, and MeFOSAA individually were associated with an increased incidence of hypertension. In the mixture analysis, women with higher overall PFAS concentrations were also more likely to develop hypertension. Taken together, our findings suggest PFAS may be an underappreciated contributing factor to women’s CVD risks.

To date, only one epidemiological study has examined the effect of PFAS on incident hypertension. In a longitudinal cohort of adults who were prediabetic and overweight in the United States with 3 follow-up visits from 1996 to 2013, Lin et al.26 did not observe an association between plasma PFAS and incident hypertension; but did report modest associations between PFOA and SBP (β=1.49 mm Hg [95% CI, 0.29–2.7] mm Hg higher per doubling in PFOA), and between MeFOSAA and prevalent hypertension (odds ratio, 1.09 [95% CI, 1.01–1.19] per doubling in MeFOSAA). PFAS concentrations in our study were similar to those in Lin et al.26 People with obesity were excluded in the Lin et al’s study26 which might account for the discrepancy between our results.

Findings from cross-sectional studies are inconsistent. Three cross-sectional studies reported significant positive associations of specific PFAS with prevalent hypertension and elevated blood pressure.27–29 In a study of 2934 adults who participated in the 2003 to 2004 and 2005 to 2006 National Health and Nutrition Examination Survey, the adjusted odds for prevalent hypertension were 2 and a half-fold higher in those at the 80th versus the 20th percentiles of PFOA concentrations; no relationship was found for other PFAS.27 A cross-sectional study of 1612 Chinese adults aged 22 to 96 years reported significant relationships of prevalent hypertension with PFOS, PFOA, perfluorononanoate, and perfluorobutanoate, with more pronounced effects observed in females; whereas no associations were detected for other compounds.28 Similarly, in a cross-sectional study of 16 224 adults aged 20 to 39 years living in Italy who were exposed to PFAS-contaminated drinking water, PFOA was associated with higher SBP in men and DBP in women; however, significant associations of PFOA, PFOS, PFHxS, and perfluorononanoate with prevalent hypertension were observed only in men.29 Other cross-sectional studies reported no associations of PFAS with prevalent hypertension and blood pressure,30–32 whereas one cross-sectional study of 154 male anglers aged 50 years and older in Wisconsin30 found an inverse association between perfluorononanoate and prevalent hypertension. All these studies considered similar sets of confounders, including demographic factors, socioeconomic status, and lifestyle factors in their analyses. It is unlikely that confounding could account for the discrepancies in the results from previous cross-sectional studies. The differences in the study results are likely attributable to differences in age. Women tend to get diagnosis with hypertension later in their midlife so that the differences in age would in differences in the study results.

A potential biological mechanism relating exposure to PFAS to hypertension may be metabolic disturbances of PFAS on fatty acid metabolism, likely through activation of multiple nuclear receptors such as PPARs.4,5 PPARα is abundantly expressed in tissues with a high capacity for mitochondrial fatty acid oxidation, such as the heart.49,50 Certain PFAS share structural similarities to fatty acids. Although human PPARα appears to be less responsive to PFAS exposure than mouse PPARα, most perfluoroalkyl carboxylates (eg, PFOA) and sulfonates (eg, PFOS) activate PPARα in mouse and human models51,52. The ability of PFAS to interfere with PPARα has been put forward as an explanation for PFAS-induced cardiovascular disturbances. Furthermore, hypertension has been associated with thyroid dysfunction, with a high prevalence in hypothyroidism.53 PFAS were associated with decreased circulating thyroid hormone levels and an interruption of the thyroid hormone signaling pathways in human studies; however, contradictory results were reported.10,54

Experimental studies have also demonstrated that PFAS exposure is related to oxidative stress and endothelial dysfunction.6,7 Oxidative stress has been proposed as a cause of hypertension. A disturbance of reactive oxygen species (ROS) production may account for reduced vasodilation, which, in turn, can trigger the development of hypertension.6 Exposure of human microvascular endothelial cells to different concentrations (10–100 μM) of PFOS in vitro increased the production of ROS and induced an alteration in endothelial permeability.7 PFOS has also been shown to induce the formation of intracellular ROS and lipid peroxidation in isolated rat hepatocytes.55 Oral administration with PFOS at 1 or 10 mg/kg bodyweight for 28 consecutive days increased intracellular ROS formation and nitric oxide production in rats, and also inhibited the activities of antioxidant enzymes (including catalase and superoxide dismutase).56 Overall, activation of nuclear receptors, interference with fatty acid metabolism, disruption of thyroid hormone homeostasis, and oxidative stress may be partly responsible for PFAS-induced elevated blood pressure and hypertension.

Notably, studies that analyzed results by sex observed a more pronounced association in women.28 Age at the final menstrual period may be a marker not only for reproductive aging but also for somatic aging. Lower levels of estrogens during the menopausal transition may increase the risks of hypertension and CVD.57 PFAS as endocrine-disrupting chemicals may accelerate ovarian aging58,59 and contribute to increased risks of hypertension and CVD during the sensitive and critical time window in a woman’s life.

We observed consistent associations of PFOS, n-PFOA, EtFOSAA, and MeFOSAA individually and PFAS mixtures with hypertension, whereas PFAS mixtures had a greater risk of incident hypertension compared to individual PFAS. Our findings are in agreement with previous toxicological studies that reported mixture effects of PFAS.60,61 Exposure to multiple PFAS is of great interest as PFAS are commonly found in drinking water, food, and a wide range of consumer and industrial products. As such, there is a pressing need to investigate how these compounds jointly affect the development of hypertension.62–66

The primary strengths of this study included prospective determination of hypertension status, a large cohort of community-based women of various racial and ethnic groups, and long follow-up of up to 18 years. The prospective design also minimized the possibility of reverse causation. Frequent follow-up visits with objective measures of blood pressure and information on hypertension medication use, instead of a 1-time questionnaire, provided reliable estimates of time to hypertension. The temporal relationship established by the long follow-up and repeated assessment of blood pressure provides evidence for the positive associations between PFAS concentrations and incident hypertension. To our knowledge, this is also the first study to examine the association of PFAS mixtures with hypertension. Our mixture analysis was powered by the well-established quantile g-computation while handling complex correlations structures among pollutants.

The findings from this study must be interpreted in light of some limitations. First, we only included midlife women. It is unknown whether these findings are applicable to men or to other women, including women of reproductive age. As described in the Methods section, the SWAN included sampling of Chinese and Japanese women at only one study site. It is possible that women at a given site may not be representative of their racial and ethnic group as a whole. In addition, we did not include Hispanic women due to lack of biologic samples. There are racial and ethnic disparities in hypertension, with higher prevalence in the Black population compared to other racial and ethnic groups.67 Causes of racial and ethnic disparities are multifactorial, and primary determinants are social factors such as education, access to healthcare, and insurance coverage.68 In SWAN, there are racial and ethnic disparities in PFAS body burden, with Black participants having higher serum concentrations of PFOS.35,36 These differences may be due to living near areas contaminated with PFAS exposure such as airports and industrial areas.69 This exposure disparity reflects the ongoing marginalization of the Black community in the United States which often lives in more environmentally vulnerable areas, reflecting decades of structural racism. Future work needs more efforts to explore these racial and ethnic differences in both exposure and outcome patterning to understand the impact of these differential exposures on minority populations. Furthermore, we did not collect information on family history of hypertension so we cannot examine effect modification by family history of hypertension. Finally, we did not consider pregnancy-related outcomes (ie, preeclampsia, gestational diabetes) in our analysis because this data was not available. No participants became pregnant during the follow-up period. We measured PFAS concentration later in life and did not have the data to assess susceptibility from early-life exposure to PFAS.

Perspectives

In a large, prospective cohort of midlife women with annual follow-up visits from 1999 to 2017, higher serum PFAS concentrations are associated with a higher risk of hypertension. Our findings suggest that PFAS are potentially modifiable risk factors for hypertension. If confirmed in future studies, these findings suggest that understanding human exposure to PFAS and developing effective strategies to reduce PFAS exposure may help prevent the development of hypertension and thereby reduce the global burden of CVD.

Article Information

Acknowledgments

The SWAN (Study of Women’s Health Across the Nation) has grant support from the National Institutes of Health (NIH), Department of Health and Human Services, through the National Institute on Aging (NIA), the National Institute of Nursing Research (NINR), and the NIH Office of Research on Women’s Health (ORWH; grants U01NR004061; U01AG012505, U01AG012535, U01AG012531, U01AG012539, U01AG012546, U01AG012553, U01AG012554, U01AG012495). The study was supported by the SWAN Repository (U01AG017719). This study was also supported by grants from the National Institute of Environmental Health Sciences (NIEHS) R01-ES026578, R01-ES026964, and P30-ES017885, and by the Center for Disease Control and Prevention (CDC)/National Institute for Occupational Safety and Health (NIOSH) grant T42-OH008455. The content of this article is solely the responsibility of the authors and does not necessarily represent the official views of the NIA, NINR, ORWH, or the NIH. The findings and conclusions of this report are those of the authors and do not necessarily represent the official position of the CDC. Use of trade names is for identification only and does not imply endorsement by the CDC, the Public Health Service, or the US Department of Health and Human Services. Clinical Centers: University of Michigan, Ann Arbor—Siobán Harlow, Principal Investigator (PI) 2011–present, MaryFran Sowers, PI 1994–2011; Massachusetts General Hospital, Boston, MA—Joel Finkelstein, PI 1999–present; Robert Neer, PI 1994–1999; Rush University, Rush University Medical Center, Chicago, IL—Howard Kravitz, PI 2009–present; Lynda Powell, PI 1994–2009; University of California, Davis/Kaiser—Ellen Gold, PI; University of California, Los Angeles—Gail Greendale, PI; Albert Einstein College of Medicine, Bronx, NY—Carol Derby, PI 2011–present, Rachel Wildman, PI 2010–2011; Nanette Santoro, PI 2004–2010; University of Medicine and Dentistry–New Jersey Medical School, Newark–Gerson Weiss, PI 1994–2004; and the University of Pittsburgh, Pittsburgh, PA—Karen Matthews, PI. NIH Program Office: National Institute on Aging, Bethesda, MD—Chhanda Dutta 2016- present; Winifred Rossi 2012–2016; Sherry Sherman 1994–2012; Marcia Ory 1994–2001; NINR, Bethesda, MD—Program Officers. Central Laboratory: University of Michigan, Ann Arbor—Daniel McConnell (Central Ligand Assay Satellite Services). CDC Laboratory: Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, GA. SWAN Repository: University of Michigan, Ann Arbor—Siobán Harlow 2013 - Present; Dan McConnell 2011 - 2013; MaryFran Sowers 2000–2011. Coordinating Center: University of Pittsburgh, Pittsburgh, PA—Maria Mori Brooks, PI 2012–present; Kim Sutton-Tyrrell, PI 2001–2012; New England Research Institutes, Watertown, MA—Sonja McKinlay, PI 1995–2001. Steering Committee: Susan Johnson, Current Chair. Chris Gallagher, Former Chair. We thank the study staff at each site and all the women who participated in SWAN.

Nonstandard Abbreviations and Acronyms

CVD

cardiovascular disease

EtFOSAA

2-(N-ethyl-perfluorooctane sulfonamido) acetate

HR

hazard ratio

MeFOSAA

2-(N-methyl-perfluorooctane sulfonamido) acetate

MPS

Multi-Pollutant Study)

N-PFOA

linear perfluorooctanoate

N-PFOS

linear perfluorooctane sulfonate

PFAS

perfluoroalkyl and polyfluoroalkyl substances

PFHxS

perfluorohexane sulfonate

PPARs

peroxisome proliferator-activated receptors

Sm-PFOS

sum of branched isomers of perfluorooctane sulfonate

Disclosures None.

Footnotes

The Data Supplement is available with this article at https://www.ahajournals.org/doi/suppl/10.1161/HYPERTENSIONAHA.121.18809.

For Sources of Funding and Disclosures, see page 1884.

Correspondence to: Sung Kyun Park, Department of Epidemiology, University of Michigan, M5541 SPH II, 1415 Washington Heights, Ann Arbor, MI 48109-2029. Email

References

  • 1. Agency for Toxic Substances and Disease Registry. Toxicological profile for trichloroethylene.; 2021. Accessed May 8, 2020. https://www.atsdr.cdc.gov/toxprofiles/tp200.pdfGoogle Scholar
  • 2. Andrews DQ, Naidenko OV. Population-wide exposure to per- and polyfluoroalkyl substances from drinking water in the united states.Environ Sci Technol Lett. 2020; 7:931–936. doi:10.1021/acs.estlett.0c00713CrossrefGoogle Scholar
  • 3. Centers for Disease Control and Prevention. Fourth National Report on Human Exposure to Environmental Chemicals.; 2021. Accessed July 5, 2021. https://www.cdc.gov/exposurereport/pdf/FourthReport_UpdatedTables_Volume2_Mar2021-508.pdfGoogle Scholar
  • 4. Bjork JA, Butenhoff JL, Wallace KB. Multiplicity of nuclear receptor activation by PFOA and PFOS in primary human and rodent hepatocytes.Toxicology. 2011; 288:8–17. doi: 10.1016/j.tox.2011.06.012CrossrefMedlineGoogle Scholar
  • 5. Hines EP, White SS, Stanko JP, Gibbs-Flournoy EA, Lau C, Fenton SE. 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.Mol Cell Endocrinol. 2009; 304:97–105. doi: 10.1016/j.mce.2009.02.021CrossrefMedlineGoogle Scholar
  • 6. Ceriello A. Possible role of oxidative stress in the pathogenesis of hypertension.Diabetes Care. 2008; 31 Suppl 2:S181–S184. doi: 10.2337/dc08-s245CrossrefMedlineGoogle Scholar
  • 7. Qian Y, Ducatman A, Ward R, Leonard S, Bukowski V, Lan Guo N, Shi X, Vallyathan V, Castranova V. Perfluorooctane sulfonate (PFOS) induces reactive oxygen species (ROS) production in human microvascular endothelial cells: role in endothelial permeability.J Toxicol Environ Health A. 2010; 73:819–836. doi: 10.1080/15287391003689317CrossrefMedlineGoogle Scholar
  • 8. Shankar A, Xiao J, Ducatman A. Perfluorooctanoic acid and cardiovascular disease in US adults.Arch Intern Med. 2012; 172:1397–1403. doi: 10.1001/archinternmed.2012.3393CrossrefMedlineGoogle Scholar
  • 9. Huang M, Jiao J, Zhuang P, Chen X, Wang J, Zhang Y. Serum polyfluoroalkyl chemicals are associated with risk of cardiovascular diseases in national US population.Environ Int. 2018; 119:37–46. doi: 10.1016/j.envint.2018.05.051CrossrefMedlineGoogle Scholar
  • 10. Kim MJ, Moon S, Oh BC, Jung D, Ji K, Choi K, Park YJ. Association between perfluoroalkyl substances exposure and thyroid function in adults: A meta-analysis.PLoS One. 2018; 13:e0197244. doi: 10.1371/journal.pone.0197244CrossrefMedlineGoogle Scholar
  • 11. Melzer D, Rice N, Depledge MH, Henley WE, Galloway TS. Association between serum perfluorooctanoic acid (PFOA) and thyroid disease in the U.S. National Health and Nutrition Examination Survey.Environ Health Perspect. 2010; 118:686–692. doi: 10.1289/ehp.0901584CrossrefMedlineGoogle Scholar
  • 12. Shrestha S, Bloom MS, Yucel R, Seegal RF, Wu Q, Kannan K, Rej R, Fitzgerald EF. Perfluoroalkyl substances and thyroid function in older adults.Environ Int. 2015; 75:206–214. doi: 10.1016/j.envint.2014.11.018CrossrefMedlineGoogle Scholar
  • 13. Winquist A, Steenland K. Modeled PFOA exposure and coronary artery disease, hypertension, and high cholesterol in community and worker cohorts.Environ Health Perspect. 2014; 122:1299–1305. doi: 10.1289/ehp.1307943CrossrefMedlineGoogle Scholar
  • 14. Lin PD, Cardenas A, Hauser R, Gold DR, Kleinman KP, Hivert MF, Fleisch AF, Calafat AM, Webster TF, Horton ES, Oken E. Per- and polyfluoroalkyl substances and blood lipid levels in pre-diabetic adults-longitudinal analysis of the diabetes prevention program outcomes study.Environ Int. 2019; 129:343–353. doi: 10.1016/j.envint.2019.05.027CrossrefMedlineGoogle Scholar
  • 15. Starling AP, Engel SM, Whitworth KW, Richardson DB, Stuebe AM, Daniels JL, Haug LS, Eggesbø M, Becher G, Sabaredzovic A, et al. Perfluoroalkyl substances and lipid concentrations in plasma during pregnancy among women in the Norwegian Mother and Child Cohort Study.Environ Int. 2014; 62:104–112. doi: 10.1016/j.envint.2013.10.004CrossrefMedlineGoogle Scholar
  • 16. Nelson JW, Hatch EE, Webster TF. Exposure to polyfluoroalkyl chemicals and cholesterol, body weight, and insulin resistance in the general U.S. population.Environ Health Perspect. 2010; 118:197–202. doi: 10.1289/ehp.0901165CrossrefMedlineGoogle Scholar
  • 17. Tian YP, Zeng XW, Bloom MS, Lin S, Wang SQ, Yim SHL, Yang M, Chu C, Gurram N, Hu LW, et al. Isomers of perfluoroalkyl substances and overweight status among chinese by sex status: isomers of C8 health project in china.Environ Int. 2019; 124:130–138. doi: 10.1016/j.envint.2019.01.006CrossrefMedlineGoogle Scholar
  • 18. Cardenas A, Hauser R, Gold DR, Kleinman KP, Hivert MF, Fleisch AF, Lin PD, Calafat AM, Webster TF, Horton ES, Oken E. Association of perfluoroalkyl and polyfluoroalkyl substances with adiposity.JAMA Netw Open. 2018; 1:e181493. doi: 10.1001/jamanetworkopen.2018.1493CrossrefMedlineGoogle Scholar
  • 19. Liu G, Dhana K, Furtado JD, Rood J, Zong G, Liang L, Qi L, Bray GA, DeJonge L, Coull B, et al. Perfluoroalkyl substances and changes in body weight and resting metabolic rate in response to weight-loss diets: a prospective study.PLoS Med. 2018; 15:e1002502. doi: 10.1371/journal.pmed.1002502CrossrefMedlineGoogle Scholar
  • 20. Ding N, Karvonen-Gutierrez CA, Herman WH, Calafat AM, Mukherjee B, Park SK. Perfluoroalkyl and polyfluoroalkyl substances and body size and composition trajectories in midlife women: the study of women’s health across the nation 1999–2018.Int J Obes. 2021; 45:1937–1948. doi:10.1038/s41366-021-00848-9CrossrefGoogle Scholar
  • 21. Lin CY, Chen PC, Lin YC, Lin LY. Association among serum perfluoroalkyl chemicals, glucose homeostasis, and metabolic syndrome in adolescents and adults.Diabetes Care. 2009; 32:702–707. doi: 10.2337/dc08-1816CrossrefMedlineGoogle Scholar
  • 22. Lind L, Zethelius B, Salihovic S, van Bavel B, Lind PM. Circulating levels of perfluoroalkyl substances and prevalent diabetes in the elderly.Diabetologia. 2014; 57:473–479. doi: 10.1007/s00125-013-3126-3CrossrefMedlineGoogle Scholar
  • 23. Cardenas A, Hivert MF, Gold DR, Hauser R, Kleinman KP, Lin PD, Fleisch AF, Calafat AM, Ye X, Webster TF, et al. Associations of perfluoroalkyl and polyfluoroalkyl substances with incident diabetes and microvascular disease.Diabetes Care. 2019; 42:1824–1832. doi: 10.2337/dc18-2254CrossrefMedlineGoogle Scholar
  • 24. Carey RM, Whelton PK; 2017 ACC/AHA Hypertension Guideline Writing Committee. Prevention, detection, evaluation, and management of high blood pressure in adults: synopsis of the 2017 american college of cardiology/american heart association hypertension guideline.Ann Intern Med. 2018; 168:351–358. doi: 10.7326/M17-3203CrossrefMedlineGoogle Scholar
  • 25. Forouzanfar MH, Liu P, Roth GA, Ng M, Biryukov S, Marczak L, Alexander L, Estep K, Hassen Abate K, Akinyemiju TF, et al. Global burden of hypertension and systolic blood pressure of at least 110 to 115 mm hg, 1990-2015.JAMA. 2017; 317:165–182. doi: 10.1001/jama.2016.19043CrossrefMedlineGoogle Scholar
  • 26. Lin PD, Cardenas A, Hauser R, Gold DR, Kleinman KP, Hivert MF, Calafat AM, Webster TF, Horton ES, Oken E. Per- and polyfluoroalkyl substances and blood pressure in pre-diabetic adults-cross-sectional and longitudinal analyses of the diabetes prevention program outcomes study.Environ Int. 2020; 137:105573. doi: 10.1016/j.envint.2020.105573CrossrefMedlineGoogle Scholar
  • 27. Min JY, Lee KJ, Park JB, Min KB. Perfluorooctanoic acid exposure is associated with elevated homocysteine and hypertension in US adults.Occup Environ Med. 2012; 69:658–662. doi: 10.1136/oemed-2011-100288CrossrefMedlineGoogle Scholar
  • 28. Bao WW, Qian ZM, Geiger SD, Liu E, Liu Y, Wang SQ, Lawrence WR, Yang BY, Hu LW, Zeng XW, Dong GH. Gender-specific associations between serum isomers of perfluoroalkyl substances and blood pressure among chinese: isomers of C8 health project in china.Sci Total Environ. 2017; 607-608:1304–1312. doi: 10.1016/j.scitotenv.2017.07.124CrossrefMedlineGoogle Scholar
  • 29. Pitter G, Zare Jeddi M, Barbieri G, Gion M, Fabricio ASC, Daprà F, Russo F, Fletcher T, Canova C. Perfluoroalkyl substances are associated with elevated blood pressure and hypertension in highly exposed young adults.Environ Health. 2020; 19:102. doi: 10.1186/s12940-020-00656-0CrossrefMedlineGoogle Scholar
  • 30. Christensen KY, Raymond M, Thompson BA, Anderson HA. Perfluoroalkyl substances in older male anglers in Wisconsin.Environ Int. 2016; 91:312–318. doi: 10.1016/j.envint.2016.03.012CrossrefMedlineGoogle Scholar
  • 31. Donat-Vargas C, Bergdahl IA, Tornevi A, Wennberg M, Sommar J, Koponen J, Kiviranta H, Åkesson A. Associations between repeated measure of plasma perfluoroalkyl substances and cardiometabolic risk factors.Environ Int. 2019; 124:58–65. doi: 10.1016/j.envint.2019.01.007CrossrefMedlineGoogle Scholar
  • 32. Geiger SD, Xiao J, Shankar A. No association between perfluoroalkyl chemicals and hypertension in children.Integr Blood Press Control. 2014; 7:1–7. doi: 10.2147/IBPC.S47660CrossrefMedlineGoogle Scholar
  • 33. El Khoudary SR, Aggarwal B, Beckie TM, Hodis HN, Johnson AE, Langer RD, Limacher MC, Manson JE, Stefanick ML, Allison MA; American Heart Association Prevention Science Committee of the Council on Epidemiology and Prevention; and Council on Cardiovascular and Stroke Nursing. Menopause transition and cardiovascular disease risk: implications for timing of early prevention: a scientific statement from the american heart association.Circulation. 2020; 142:e506–e532. doi: 10.1161/CIR.0000000000000912LinkGoogle Scholar
  • 34. Santoro N, Sutton-Tyrrell K. The SWAN song: study of women’s health across the nation’s recurring themes.Obstet Gynecol Clin North Am. 2011; 38:417–423. doi: 10.1016/j.ogc.2011.05.001CrossrefMedlineGoogle Scholar
  • 35. Park SK, Peng Q, Ding N, Mukherjee B, Harlow SD. Determinants of per- and polyfluoroalkyl substances (PFAS) in midlife women: evidence of racial/ethnic and geographic differences in pfas exposure.Environ Res. 2019; 175:186–199. doi: 10.1016/j.envres.2019.05.028CrossrefMedlineGoogle Scholar
  • 36. Ding N, Harlow SD, Batterman S, Mukherjee B, Park SK. Longitudinal trends in perfluoroalkyl and polyfluoroalkyl substances among multiethnic midlife women from 1999 to 2011: the study of women’s health across the nation.Environ Int. 2020; 135:105381. doi: 10.1016/j.envint.2019.105381CrossrefMedlineGoogle Scholar
  • 37. Wang X, Mukherjee B, Batterman S, Harlow SD, Park SK. Urinary metals and metal mixtures in midlife women: the study of women’s health across the nation (swan).Int J Hyg Environ Health. 2019; 222:778–789. doi: 10.1016/j.ijheh.2019.05.002CrossrefMedlineGoogle Scholar
  • 38. Carey RM, Cutler J, Friedewald W, et al. The 1984 report of the joint national committee on detection, evaluation and treatment of high blood pressure.Nurse Pract. 1985; 10:9–33. doi:10.1097/00006205-198507000-00003CrossrefMedlineGoogle Scholar
  • 39. Chobanian AV, Bakris GL, Black HR, Cushman WC, Green LA, Izzo JL, Jones DW, Materson BJ, Oparil S, Wright JT, Roccella EJ; National Heart, Lung, and Blood Institute Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure; National High Blood Pressure Education Program Coordinating Committee. The Seventh report of the joint national committee on prevention, detection, evaluation, and treatment of high blood pressure: the JNC 7 report.JAMA. 2003; 289:2560–2572. doi: 10.1001/jama.289.19.2560CrossrefMedlineGoogle Scholar
  • 40. Kato K, Basden BJ, Needham LL, Calafat AM. Improved selectivity for the analysis of maternal serum and cord serum for polyfluoroalkyl chemicals.J Chromatogr A. 2011; 1218:2133–2137. doi: 10.1016/j.chroma.2010.10.051CrossrefMedlineGoogle Scholar
  • 41. Hornung RW, Reed LD. Estimation of Average Concentration in the Presence of Nondetectable Values.Appl Occup Environ Hyg. 1990; 5:46–51. doi:10.1080/1047322X.1990.10389587CrossrefGoogle Scholar
  • 42. Wassertheil-Smoller S, Anderson G, Psaty BM, Black HR, Manson J, Wong N, Francis J, Grimm R, Kotchen T, Langer R, Lasser N. Hypertension and its treatment in postmenopausal women: baseline data from the Women’s Health Initiative.Hypertension. 2000; 36:780–789. doi: 10.1161/01.hyp.36.5.780LinkGoogle Scholar
  • 43. Coghlin J, Hammond SK, Gann PH. Development of epidemiologic tools for measuring environmental tobacco smoke exposure.Am J Epidemiol. 1989; 130:696–704. doi: 10.1093/oxfordjournals.aje.a115391CrossrefMedlineGoogle Scholar
  • 44. Sternfeld B, Ainsworth BE, Quesenberry CP. Physical activity patterns in a diverse population of women.Prev Med. 1999; 28:313–323. doi: 10.1006/pmed.1998.0470CrossrefMedlineGoogle Scholar
  • 45. Thiébaut AC, Bénichou J. Choice of time-scale in cox’s model analysis of epidemiologic cohort data: a simulation study.Stat Med. 2004; 23:3803–3820. doi: 10.1002/sim.2098CrossrefMedlineGoogle Scholar
  • 46. Keil AP, Buckley JP, O’Brien KM, Ferguson KK, Zhao S, White AJ. A quantile-based g-computation approach to addressing the effects of exposure mixtures.Environ Health Perspect. 2020; 128:47004. doi: 10.1289/EHP5838CrossrefMedlineGoogle Scholar
  • 47. Carrico C, Gennings C, Wheeler DC, Factor-Litvak P. Characterization of weighted quantile sum regression for highly correlated data in a risk analysis setting.J Agric Biol Environ Stat. 2015; 20:100–120. doi: 10.1007/s13253-014-0180-3CrossrefMedlineGoogle Scholar
  • 48. Ding N, Karvonen-Gutierrez CA, Herman WH, Calafat AM, Mukherjee B, Park SK. Associations of perfluoroalkyl and polyfluoroalkyl substances (PFAS) and PFAS mixtures with adipokines in midlife women.Int J Hyg Environ Health. 2021; 235:113777. doi: 10.1016/j.ijheh.2021.113777CrossrefMedlineGoogle Scholar
  • 49. Barger PM, Kelly DP. PPAR signaling in the control of cardiac energy metabolism.Trends Cardiovasc Med. 2000; 10:238–245. doi: 10.1016/s1050-1738(00)00077-3CrossrefMedlineGoogle Scholar
  • 50. Gilde AJ, van der Lee KA, Willemsen PH, Chinetti G, van der Leij FR, van der Vusse GJ, Staels B, van Bilsen M. Peroxisome proliferator-activated receptor (PPAR) alpha and PPARbeta/delta, but not PPARgamma, modulate the expression of genes involved in cardiac lipid metabolism.Circ Res. 2003; 92:518–524. doi: 10.1161/01.RES.0000060700.55247.7CLinkGoogle Scholar
  • 51. Wolf CJ, Takacs ML, Schmid JE, Lau C, Abbott BD. Activation of mouse and human peroxisome proliferator-activated receptor alpha by perfluoroalkyl acids of different functional groups and chain lengths.Toxicol Sci. 2008; 106:162–171. doi: 10.1093/toxsci/kfn166CrossrefMedlineGoogle Scholar
  • 52. Takacs ML, Abbott BD. Activation of mouse and human peroxisome proliferator-activated receptors (alpha, beta/delta, gamma) by perfluorooctanoic acid and perfluorooctane sulfonate.Toxicol Sci. 2007; 95:108–117. doi: 10.1093/toxsci/kfl135CrossrefMedlineGoogle Scholar
  • 53. Danzi S, Klein I. Thyroid hormone and blood pressure regulation.Curr Hypertens Rep. 2003; 5:513–520. doi: 10.1007/s11906-003-0060-7CrossrefMedlineGoogle Scholar
  • 54. Ballesteros V, Costa O, Iñiguez C, Fletcher T, Ballester F, Lopez-Espinosa MJ. Exposure to perfluoroalkyl substances and thyroid function in pregnant women and children: a systematic review of epidemiologic studies.Environ Int. 2017; 99:15–28. doi: 10.1016/j.envint.2016.10.015CrossrefMedlineGoogle Scholar
  • 55. Khansari MR, Yousefsani BS, Kobarfard F, Faizi M, Pourahmad J. In vitro toxicity of perfluorooctane sulfonate on rat liver hepatocytes: probability of distructive binding to CYP 2E1 and involvement of cellular proteolysis.Environ Sci Pollut Res Int. 2017; 24:23382–23388. doi: 10.1007/s11356-017-9908-2CrossrefMedlineGoogle Scholar
  • 56. Han R, Hu M, Zhong Q, Wan C, Liu L, Li F, Zhang F, Ding W. Perfluorooctane sulphonate induces oxidative hepatic damage via mitochondria-dependent and NF-κB/TNF-α-mediated pathway.Chemosphere. 2018; 191:1056–1064. doi: 10.1016/j.chemosphere.2017.08.070CrossrefMedlineGoogle Scholar
  • 57. Zhao D, Guallar E, Ouyang P, Subramanya V, Vaidya D, Ndumele CE, Lima JA, Allison MA, Shah SJ, Bertoni AG, et al. Endogenous sex hormones and incident cardiovascular disease in post-menopausal women.J Am Coll Cardiol. 2018; 71:2555–2566. doi: 10.1016/j.jacc.2018.01.083CrossrefMedlineGoogle Scholar
  • 58. Ding N, Harlow SD, Randolph JF, Calafat AM, Mukherjee B, Batterman S, Gold EB, Park SK. Associations of perfluoroalkyl substances with incident natural menopause: the study of women’s health across the nation.J Clin Endocrinol Metab. 2020; 105:dgaa303. doi: 10.1210/clinem/dgaa303CrossrefMedlineGoogle Scholar
  • 59. Harlow SD, Hood MM, Ding N, Mukherjee B, Calafat AM, Randolph JF, Gold EB, Park SK. Per- and polyfluoroalkyl substances and hormone levels during the menopausal transition.J Clin Endocrinol Metab. 2021; 106:e4427–e4437. doi: 10.1210/clinem/dgab476CrossrefMedlineGoogle Scholar
  • 60. G D, J Z, Y C, et al. Combined effects of PFOS and PFOA on zebrafish (danio rerio) embryos.Arch Environ Contam Toxicol. 2013; 64:668–675. doi:10.1007/S00244-012-9864-2CrossrefMedlineGoogle Scholar
  • 61. Ojo AF, Peng C, Ng JC. Combined effects and toxicological interactions of perfluoroalkyl and polyfluoroalkyl substances mixtures in human liver cells (HepG2).Environ Pollut. 2020; 263(Pt B):114182. doi: 10.1016/j.envpol.2020.114182CrossrefMedlineGoogle Scholar
  • 62. Wang X, Karvonen-Gutierrez CA, Herman WH, Mukherjee B, Harlow SD, Park SK. Urinary metals and incident diabetes in midlife women: study of women’s health across the nation (SWAN).BMJ Open Diabetes Res Care. 2020; 8:e001233. doi: 10.1136/bmjdrc-2020-001233CrossrefMedlineGoogle Scholar
  • 63. Wang X, Mukherjee B, Karvonen-Gutierrez CA, Herman WH, Batterman S, Harlow SD, Park SK. Urinary metal mixtures and longitudinal changes in glucose homeostasis: The study of women’s health across the nation (SWAN).Environ Int. 2020; 145:106109. doi: 10.1016/j.envint.2020.106109CrossrefMedlineGoogle Scholar
  • 64. Wang X, Karvonen-Gutierrez CA, Herman WH, Mukherjee B, Harlow SD, Park SK. Urinary heavy metals and longitudinal changes in blood pressure in midlife women: the study of women’s health across the nation.Hypertension. 2021; 78:543–551. doi: 10.1161/HYPERTENSIONAHA.121.17295LinkGoogle Scholar
  • 65. Wang X, Mukherjee B, Park SK. Associations of cumulative exposure to heavy metal mixtures with obesity and its comorbidities among U.S. adults in NHANES 2003-2014.Environ Int. 2018; 121(pt 1):683–694. doi: 10.1016/j.envint.2018.09.035CrossrefMedlineGoogle Scholar
  • 66. Wang X, Mukherjee B, Park SK. Does information on blood heavy metals improve cardiovascular mortality prediction?J Am Heart Assoc. 2019; 8:e013571. doi: 10.1161/JAHA.119.013571LinkGoogle Scholar
  • 67. Whelton PK, Carey RM, Aronow WS, Casey DE, Collins KJ, Dennison Himmelfarb C, DePalma SM, Gidding S, Jamerson KA, Jones DW, et al. 2017 ACC/AHA/AAPA/ABC/ACPM/AGS/APhA/ASH/ASPC/NMA/PCNA guideline for the prevention, detection, evaluation, and management of high blood pressure in adults: a report of the american college of cardiology/american heart association task force on clinical practice guidelines.Hypertension. 2018; 71:e13–e115. doi: 10.1161/HYP.0000000000000065LinkGoogle Scholar
  • 68. Gu A, Yue Y, Desai RP, Argulian E. Racial and ethnic differences in antihypertensive medication use and blood pressure control among us adults with hypertension: the national health and nutrition examination survey, 2003 to 2012.Circ Cardiovasc Qual Outcomes. 2017; 10:e003166. doi: 10.1161/CIRCOUTCOMES.116.003166LinkGoogle Scholar
  • 69. Hu XC, Andrews DQ, Lindstrom AB, Bruton TA, Schaider LA, Grandjean P, Lohmann R, Carignan CC, Blum A, Balan SA, et al. Detection of poly- and perfluoroalkyl substances (PFASs) in U.S. drinking water linked to industrial Sites, Military Fire Training Areas, and Wastewater Treatment Plants.Environ Sci Technol Lett. 2016; 3:344–350. doi: 10.1021/acs.estlett.6b00260CrossrefMedlineGoogle Scholar

eLetters(0)

eLetters should relate to an article recently published in the journal and are not a forum for providing unpublished data. Comments are reviewed for appropriate use of tone and language. Comments are not peer-reviewed. Acceptable comments are posted to the journal website only. Comments are not published in an issue and are not indexed in PubMed. Comments should be no longer than 500 words and will only be posted online. References are limited to 10. Authors of the article cited in the comment will be invited to reply, as appropriate.

Comments and feedback on AHA/ASA Scientific Statements and Guidelines should be directed to the AHA/ASA Manuscript Oversight Committee via its Correspondence page.