You are on page 1of 10

Environment International 156 (2021) 106599

Contents lists available at ScienceDirect

Environment International
journal homepage: www.elsevier.com/locate/envint

Perfluoroalkyl substances and immune cell counts in adults from the


Mid-Ohio Valley (USA)
Maria-Jose Lopez-Espinosa a, b, c, *, Christian Carrizosa a, Michael I. Luster d, Joseph
B. Margolick e, Olga Costa a, Giovanni S. Leonardi f, g, Tony Fletcher f, g
a
Epidemiology and Environmental Health Joint Research Unit, FISABIO− Universitat Jaume I− Universitat de València, 46020 Valencia, Spain
b
Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain
c
Faculty of Nursing and Chiropody, Universitat de València, 46001 Valencia, Spain
d
School of Public Health, West Virginia University, Morgantown, WV 26505, USA
e
Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21279, USA
f
Centre for Radiation, Chemical and Environmental Hazards, Public Health England (PHE), Chilton, Oxfordshire OX11 0RQ, United Kingdom
g
Department of Public Health, Environments and Society, London School of Hygiene and Tropical Medicine, London WCIH 9SH, United Kingdom

A R T I C L E I N F O A B S T R A C T

Handling Editor: Shoji F. Nakayama Background: Although perfluoroalkyl substances (PFASs) may be immunotoxic, evidence for this in humans is
scarce. We studied the association between 4 PFASs (perfluorohexane sulfonate [PFHxS], perfluorooctanoic acid
Keywords: [PFOA], perfluorooctane sulfonate [PFOS] and perfluorononanoic acid [PFNA]) and circulating levels of several
PFASs types of immune cells.
Immune system
Methods: Serum PFASs and white blood cell types were measured in 42,782 (2005–2006) and 526 (2010) adults
White blood cells
from an area with PFOA drinking water contamination in the Mid-Ohio Valley (USA). Additionally, the major
Lymphocytes
lymphocyte subsets were measured in 2010. Ln(cell counts) and percentages of cell counts were regressed on
serum PFAS concentrations (ln or percentiles). Adjusted results were expressed as the percentage difference (95%
CI) per interquartile range (IQR) increment of each PFAS concentration.
Results: Generally positive monotonic associations between total lymphocytes and PFHxS, PFOA, and PFOS were
found in both surveys (difference range: 1.12–7.33% for count and 0.36–1.77 for percentage, per PFAS IQR
increment), and were stronger for PFHxS. These associations were reflected in lymphocyte subset counts but not
percentages, with PFHxS positively and monotonically associated with T, B, and natural killer (NK) cell counts
(range: 5.51–8.62%), PFOA and PFOS with some T-cell phenotypes, and PFOS with NK cells (range:
3.12–12.21%), the associations being monotonic in some cases. Neutrophils, particularly percentage (range:
− 1.74 to − 0.36), showed decreasing trends associated with PFASs. Findings were less consistent for monocytes
and eosinophils.
Conclusion: These results suggest an association between PFHxS and, less consistently, for PFOA and PFOS, and
total lymphocytes (although the magnitudes of the differences were small). The increase in absolute lymphocyte
count appeared to be evenly distributed across lymphocyte subsets since associations with their percentages were
not significant.

1. Introduction (approximately 4–6 years) (Agency for Toxic Substances and Disease
Registry [ATSDR], 2018). Whereas mean serum concentrations of PFASs
Perfluoroalkyl substances (PFASs) are a diverse group of chemicals have decreased in recent years in Western countries (Sunderland et al.,
used in industrial applications and consumer products since the 1950s. 2019), probably because their production has been reduced or has been
They persist in the environment, resist degradation, bioaccumulate in banned (ATSDR, 2018), the stability of these compounds and recent
humans, and have relatively long half-lives in the body of humans increases in serum levels observed in Asia and the Middle East

* Corresponding author at: Foundation for the Promotion of Health and Biomedical Research in the Valencian Region, FISABIO-Public Health, Avenida de Cat­
alunya 21, 46020 Valencia, Spain.
E-mail address: lopez_josesp@gva.es (M.-J. Lopez-Espinosa).

https://doi.org/10.1016/j.envint.2021.106599
Received 22 December 2020; Received in revised form 18 March 2021; Accepted 21 April 2021
Available online 13 May 2021
0160-4120/© 2021 The Authors. Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
M.-J. Lopez-Espinosa et al. Environment International 156 (2021) 106599

(International Pollutants Elimination Network, 2019) mean that PFASs, 2. Methods


and their possible health effects, remain significant public health
concerns. 2.1. Study design and subjects
To date, most of the animal and human studies on the health effects
of some PFASs have focused on perfluorooctanoic acid (PFOA) and The C8 Health Project arose from the settlement of a class action
perfluorooctane sulfonate (PFOS), and to a lesser extent on per­ lawsuit filed in 2002 by residents living near the DuPont Washington
fluorohexane sulfonate (PFHxS) and perfluorononanoic acid (PFNA). Works plant in West Virginia regarding the release of PFOA from the
The results of those studies suggest that they may affect metabolic, facility that had contaminated drinking water supplies along the Mid-
endocrine, and immune processes (Ballesteros et al., 2017; EFSA [Eu­ Ohio River Valley. The project collected data on 69,030 subjects (esti­
ropean Food Safety Authority], 2018; Lau et al., 2007; Matilla- mated participation rate for residents at the time: 80%) between August
Santander et al., 2017; United States Department of Health and 2005 and August 2006. Individuals were eligible for the study if they
Human Services-National Toxicology Program [USDHHS-NTP], 2016). had consumed water for at least one year between 1950 and 2004 from
Regarding the immune system, an extensive review of the literature by any of the six contaminated water districts or from private wells in the
the United States National Toxicology Program (USDHHS-NTP, 2016) proximity of the chemical plant. Details of the study population have
concluded that both PFOA and PFOS are immunotoxic. This conclusion been described elsewhere (Frisbee et al., 2009).
was based primarily upon a high level of evidence from animal studies Between March and July 2010, participants were recalled and
showing that at high concentrations they suppress the antibody invited to participate in a second survey, as described elsewhere (Fitz-
response, and a moderate level of evidence from human studies indi­ Simon et al., 2013). Briefly, the second survey was restricted to residents
cating that they affect vaccine responses. This conclusion is also sup­ of the three Ohio and five West Virginia ZIP code areas in the region
ported by a more recent review from the European Food Safety most affected by PFOA contamination. The follow-up population was
Authority (EFSA), which reported an adverse effect on serum antibody restricted to those aged 20–59 at the time of the initial survey
response after vaccination due to PFOS (and possibly PFOA) exposure, (2005–2006), and individuals were chosen randomly from age–gender
with children considered to be the most vulnerable group (EFSA, 2018). strata to give balanced numbers of men and women in each ten-year age
Regarding PFHxS and PFNA exposure, there is still insufficient epide­ group. Altogether 973 people were contacted, and after excluding peo­
miological data on their possible immunotoxicity (Chen et al., 2018; ple who reported a past or present diagnosis of cancer and those un­
Dong et al., 2013; Grandjean et al., 2017) to reach a conclusion. willing to provide blood samples, 748 participated, with ages between
Most observational studies in humans have investigated associations 24 and 64 years at the time of the second survey. From the first survey
between PFASs and immune outcomes such as asthma, allergies, pro­ containing participants of all ages, we focused on the subset of adults
pensity to infections, and serum antibody response to vaccination aged 24–64 years at baseline (2005–2006) so as to be in the same age
(EFSA, 2018). However, immunotoxicity can also be studied by range as those in the follow-up study (2010).
measuring peripheral counts of white blood cells (WBCs: neutrophils, Information on infections was not available in the first survey, but
lymphocytes, monocytes, eosinophils, and basophils) and subsets of this information was available in 2010. Participants were asked if, at the
peripheral blood lymphocytes delineated by cell-surface markers time of the survey, they had any of the following infections or conditions
(CD3+T cells, CD3+CD4+T helper cells, CD3+CD8+T-cytotoxic cells, associated with inflammation: cold, influenza, sore throat, bronchitis,
CD3+CD4+ CD8+ double positive [DP] T cells, CD3− CD16+ CD56+ pneumonia, gastroenteritis, cold sore, shingles, and/or sinus, ear, tooth,
natural killer [NK] cells, CD3− CD19+B cells, and CD4+/CD8+ ratio). and/or mouth infections. A total of 213 out of 748 participants reported
WBC count and peripheral blood lymphocyte measurements are at least one of the above-mentioned conditions, with sinus and skin in­
powerful diagnostic tools used primarily in the identification and pro­ fections being the most common. Analyses for the follow-up study
gression of diseases such as acquired immunodeficiency syndrome focused on those who reported not having any infections or conditions
(AIDS), and leukemias and lymphomas (Castilho et al., 2016; Davis associated with inflammation plus PFAS analyses. We chose to exclude
et al., 2014). They have also been used occasionally in clinical studies for those with infections as they could have an effect on immune cell counts
testing both the efficacy and the toxicity of therapeutics (e.g., Aziz et al., (George-Gay and Parker, 2003). Altogether, 42,782 and 526 adults in
2013), and to help identify immunotoxicity following environmental the first and second surveys, respectively, gave a blood sample, had
chemical exposure (Haase et al., 2016; Leonardi et al., 2000; Tryphonas, analyses of both PFASs and immune markers (2005–2006: types of
2001). Nevertheless, data obtained from human epidemiologic studies WBCs, and 2010: types of WBCs and subtypes of lymphocytes), as well as
on PFAS exposure and these immune markers is still limited, with some data on sociodemographic and anthropometric factors, and were
data derived from monitoring humans exposed to PFASs in the work­ included in the present study. The London School of Hygiene and
place (Costa et al., 2009; Olsen et al., 2003) or from cross-sectional Tropical Medicine Ethics Committee approved this study and informed
(Abraham et al., 2020; Dong et al., 2013; Emmett et al., 2006; Gay­ consent was obtained from all participants in each period.
lord et al., 2019; Knudsen et al., 2018) or longitudinal (Oulhote et al.,
2017) non-occupational human studies. Furthermore, only a limited 2.2. Serum PFAS concentrations
number of immunological end points have been studied in most of the
previous studies (predominantly, total WBCs, and/or eosinophils). Laboratory analyses of serum PFASs were conducted by Exygen,
Given these limitations, further studies are warranted. State College, PA, USA (a commercial laboratory) in 2005–2006 and by
Previous work by our group showed that PFOA may be associated the Centers for Disease Control and Prevention (CDC) in Atlanta, GA,
with a decrease in influenza vaccine efficacy in adults who had had an USA in 2010. The analytical (Flaherty et al., 2005; Frisbee et al., 2009;
A/H3N2 seasonal flu vaccination (Looker et al., 2014). To further Kato et al., 2011) and quality control (Van Leeuwen et al., 2006) pro­
address immunotoxicity, we aimed to evaluate the association between cedures employed by both laboratories have been described elsewhere.
PFAS concentrations and WBC counts (measured in both 2005–2006 and Briefly, laboratory analyses of PFASs used online solid phase extraction
2010), and the major lymphocyte subsets (measured in 2010) amongst coupled with reversed-phase high-performance liquid chromatography
adults who had been highly exposed to PFOA due to contaminated water separation and detection by isotope-dilution tandem mass spectrometry.
supplies, but with similar exposure to other PFASs compared to other The limits of detection (LODs) in 2005–2006 were 0.5 ng/mL for all
populations from North America (Frisbee et al., 2009). PFASs. In 2010, LODs were 0.1 ng/mL for PFNA and PFHxS, 0.2 ng/mL
for PFOS, and 0.5 ng/mL for PFOA.

2
M.-J. Lopez-Espinosa et al. Environment International 156 (2021) 106599

2.3. Immune cell analysis survey). We also excluded from the models those participants with self-
reported autoimmune diseases (n = 1,606 in 2005–2006, n = 28 in
Whole blood collected in EDTA vacutainer tubes was shipped to 2010) to account for medical conditions that could affect immunological
laboratories for analysis of immune cells. In LabCorp Inc., Burlington, measurements (specifically, systemic lupus erythematosus, scleroderma,
NC, USA (an accredited clinical diagnostic laboratory), analyses of types multiple sclerosis, Sjögren’s syndrome, Addison’s disease, and rheu­
of WBCs (neutrophils, monocytes, eosinophils, lymphocytes, and baso­ matoid arthritis) and those who reported cancer (n = 2,614 in the
phils) were performed using an automated hematology analyzer (LH 2005–2006 survey, since in the follow-up study participants with this
Series, Beckman Coulter). In the flow cytometry core laboratory at Johns condition were not included in the study). Although the main analyses in
Hopkins Bloomberg School of Public Health in Baltimore, immunophe­ 2010 were restricted to participants who reported no infection at the
notypic analysis was performed on peripheral blood lymphocytes time of the interview (n = 526), a sensitivity analysis was performed on
(CD3+ T cells, CD3+ CD4+ T helper cells, CD3+ CD8+ T-cytotoxic cells, the full population (n = 736) repeating the main analyses. Finally, we
CD3+ CD4+ CD8+ DP T cells, CD3− CD16+ CD56+ NK cells, CD3− also investigated possible differences by gender by including the inter­
CD19+ B cells) using standard flow cytometry methods, as previously action term with the contaminants in the models.
described (Giorgi et al., 1990; Schenker et al., 1993). The CD4+/CD8+ We expressed results as percentage difference (with a 95% confi­
ratio was also calculated. The laboratory was certified by two profi­ dence interval – CI) in the outcomes associated with an interquartile
ciency testing programs, i.e., the Immunology Quality Assurance (IQA) range (IQR) increment in PFAS concentrations. The percentage differ­
administered by the US National Institutes of Health, and the College of ence was calculated as the complement of the exponentiated regression
American Pathologists (CAP) program. The laboratory was also certified coefficient [(exp(β × IQR) – 1) × 100] for cell counts and just the
under the provisions of the Clinical Laboratory Improvement Act (CLIA). regression coefficient (β × IQR) for cell percentages. We used the
2005–2006 IQRs of ln(PFASs) for both populations to make results
2.4. Covariates comparable. For easier clinical interpretation, results for count data
were also expressed in terms of absolute differences, taking the median
We obtained information on sociodemographic and anthropometric values as the reference [median(cells) * (exp(β × IQR) – 1)]. Differences
characteristics, as well as on lifestyle variables, through questionnaires in percentages of cells were evaluated to determine whether there were
that were administered by trained interviewers in person (2005–2006) differential effects of PFASs on this relative scale as opposed to the ef­
and by telephone (2010). We considered the following variables: fects on absolute counts (e.g., an increase in total lymphocyte count
gender, race/ethnicity, age, education, body mass index (BMI, only might not occur evenly across the different subsets).
available in the 2005–2006 survey), tobacco consumption, alcohol We also re-ran the main analysis with deciles (2005–2006 popula­
intake, present or past diagnosis of immune diseases and/or cancer, and tion) and tertiles (2010 population) of PFASs to explore the shape of the
anti-inflammatory medication (only available for the 2010 survey). The exposure–response curve. To be consistent with the regression on the ln
participants were asked if they were taking or had taken in the last four (PFASs) scale, tests for trend were conducted across percentiles
years paracetamol, aspirin, or other anti-inflammatory medications on a including the ln-transformed geometric mean of each decile or tertile
regular basis (i.e., at least once a week for six months or more). added to the models as a numerical regressor. Results of categorical
Serum creatinine levels were determined using the Kinetic Jaffe analyses are presented graphically as the adjusted value of each
method (LabCorp, Burlington, NC). We estimated glomerular filtration outcome, by exposure group, with 95% CI.
rate (eGFR) using the Modification of Diet in Renal Disease (MDRD) To be conservative, we used heteroscedasticity-consistent standard
Study 4-variable equation (Levey et al., 2006). errors in all the analyses to account for potential deviations from the
linear model assumptions (Long and Ervin, 2000). We conducted sta­
2.5. Statistical analysis tistical analyses using R.3.6.2 (R Core Team, 2020).

We performed statistical analyses for all immune cell types except 3. Results
basophils (due to the low proportion of samples with these immune cells
present: 25.6% in 2005–2006 and 17.9% in 2010). For PFASs, the % 3.1. Study population, immune outcomes, and PFAS concentrations
≤LOD was very low (<0.1% for PFOA up to 2.6% for PFHxS) and they
were set to half the LOD for the analyses. Pearson’s correlations between The baseline population (2005–2006) was comprised of similar
the four ln(PFASs), and these contaminants and eGFR were performed, numbers of women (53%) and men (47%), 29% of whom were smokers,
and a heatmap was also generated to display them. We used ordinal and 72% were overweight or obese. Significant differences between the
mixed models to assess the baseline to follow-up differences in socio­ 2005–2006 and 2010 surveys were found in some characteristics, with
demographic characteristics. Linear mixed models adjusted for cova­ higher educational levels, fewer smokers and alcohol drinkers, and
riates were also employed to study the changes in exposure levels of ln slightly older participants in 2010 (n = 526, mean age: 46 years)
(PFASs) over time. compared to 2005–2006 (n = 42,782, mean age: 44 years) (Table 1).
We used multivariate linear regression analysis to assess the relation PFHxS, PFOA, PFOS, and PFNA were detectable (above the LOD) in
between ln(counts) and percentages of immune cells and ln(PFASs) at 97.4, 99.9, 99.5, and 97.6% of people in 2005–2006. Corresponding
the baseline (n = 42,782) and follow-up (n = 526). All models were data for 2010 were: 99.4, 99.8, 99.4, and 99.2% (data not shown in
adjusted for gender, age in 10-year categories, smoking, month of table). The median serum PFASs in the follow-up compared to baseline
sampling (because of a decreasing trend in serum PFASs during the was higher for PFOA (due to the study population being located in the
collection period in this population), alcohol intake, and educational six most contaminated water districts in 2010), lower for PFOS, similar
level (hereafter called ‘main analysis’). for PFHxS, and the same for PFNA (Table 1). In 2005–2006, the IQR
We also conducted several sensitivity analyses including: BMI (since contrast for PFOA was bigger (56.0 ng/ml), PFOS was intermediate
excess adiposity has been related to both impaired immune function (15.1 ng/ml), and both PFHxS and PFNA were smaller (2.8 and 0.7 ng/
(Martí et al., 2001) and PFASs (Braun, 2017; Cardenas et al., 2018), we ml, respectively). IQR contrasts for 2010 were: 69.8, 9.0, 2.0, and 0.7
preferred not to include this variable in the main analysis); the four ng/mL for PFOA, PFOS, PFHxS, and PFNA, respectively (data not shown
PFASs simultaneously; eGFR (to evaluate whether a common disease in table). Furthermore, positive correlations (p < 0.001 in all cases) were
process may be contributing to altered PFAS excretion from the kidney found between all the PFASs analyzed (r range: 0.32–0.57 in 2005–2006
and increasing immune cell counts) (Dhingra et al., 2017; Watkins et al., and 0.44–0.77 in 2010). We found weak inverse correlations between
2013); and use of anti-inflammatory drugs (only available in the 2010 PFASs and eGFR (r range from − 0.13 to − 0.03, with p < 0.05 except for

3
M.-J. Lopez-Espinosa et al. Environment International 156 (2021) 106599

Table 1 Table 2
Study population and PFAS concentrations, Mid-Ohio Valley, USA (2005–2010). Counts of WBCs and lymphocyte subsets, and association between PFASs and
Variable 2005–2006 2010
relative (percent) differences in these counts amongst the population in
2005–2006 (n = 42,782) and in 2010 (n = 526), Mid-Ohio Valley, USA.
(n ¼ 42,782) (n ¼ 526) pa
Sex 0.292 Cell PFHxS PFOA PFOS PFNA
Female 22,542 (52.7) 265 (50.4) count
Male 20,240 (47.3) 261 (49.6) Outcome Median % diff. % diff. % diff. % diff.
Race/ethnicity 0.071 (IQR) (95% (95% (95% (95%
Non-Hispanic white 41,735 (97.6) 519 (98.7) CI) CI) CI) CI)
Others 1,047 (2.45) 7 (1.33) Total WBCs 7,100 0.55 − 0.27 − 0.55 − 0.21
Age <0.001 2005–2006 (5,900, (0.25, (-0.62, (-0.84, (-0.47,
≤30 years 7,745 (18.1) 62 (11.8) 8,600) 0.86) 0.08) − 0.26) 0.05)
31–40 years 10,444 (24.4) 122 (23.2) Total WBCs 2010 6,300 0.90 0.84 0.55 − 0.08
41–50 years 11,804 (27.6) 132 (25.1) (5,300, (-1.47, (-2.20, (-1.35, (-1.95,
51–60 years 9,808 (22.9) 138 (26.2) 7,600) 3.33) 3.97) 2.49) 1.82)
>60 years 2,981 (6.97) 72 (13.7) Neutrophils 4,400 − 0.04 − 0.92 − 1.56 − 0.77
Education <0.001 2005–2006 (3,500, (-0.44, (-1.37, (-1.93, (-1.11,
<12 years 3,902 (9.12) 16 (3.04) 5,500) 0.35) − 0.47) − 1.19) − 0.44)
HSD or GED 17,461 (40.8) 158 (30.0) Neutrophils 2010 4,000 − 1.99 − 1.44 − 0.86 − 1.14
Some college 14,881 (34.8) 242 (46.0) (3,200, (-4.97, (-5.12, (-3.38, (-3.68,
≥Bachelor’s degree 6,538 (15.3) 110 (20.9) 5,000) 1.08) 2.39) 1.72) 1.46)
BMI 0.436 Monocytes 400 1.16 0.61 0.38 0.15
Underweight 486 (1.14) 6 (1.14) 2005–2006 (300, (0.71, (0.08, (-0.06, (-0.24,
Normal weight 11,607 (27.1) 139 (26.4) 500) 1.63) 1.15) 0.82) 0.54)
Overweight 14,826 (34.7) 206 (39.2) Monocytes 2010 300 1.85 4.29 1.41 0.55
Obese I 9,198 (21.5) 96 (18.3) (300, (-2.71, (-1.05, (-2.38, (-2.81,
Obese II and III 6,665 (15.6) 79 (15.0) 400) 6.63) 9.92) 5.35) 4.02)
Smoking <0.001 Eosinophils 100 − 0.96 − 0.06 − 0.29 − 0.76
Never smoked 20,017 (46.8) 331 (62.9) 2005–2006 (100, (-1.76, (-0.98, (-1.08, (-1.44,
Ex-smoker 10,509 (24.6) 111 (21.1) 200) − 0.15) 0.87) 0.50) − 0.07)
Current smoker 12,256 (28.6) 84 (16.0) Eosinophils 2010 100 1.15 4.77 4.22 6.37
Alcohol intake <0.001 (100, (-6.64, (-4.11, (-2.33, (0.68,
Lifetime non-drinker 7,898 (18.5) 303 (57.6) 200) 9.59) 14.46) 11.21) 12.38)
Ex-drinker 12,207 (28.5) 52 (9.89) Lymphocytes 2,000 2.00 1.12 1.95 1.47
Drinker 22,677 (53.0) 171 (32.5) 2005–2006 (1,700, (1.63, (0.70, (1.57, (1.15,
PFAS (ng/mL) 2,500) 2.37) 1.55) 2.33) 1.79)
PFHxS 2.90(1.80, 4.60) 2.10(1.20, 3.20) <0.001 Lymphocytes 2010 1,800 7.33 5.50 3.39 1.29
PFOA 26.9(13.2, 69.2) 35.7(15.0, 93.7) <0.001 (1,500, (4.12, (1.52, (0.70, (-1.04,
PFOS 19.7(13.3, 28.4) 9.60(6.10, 14.9) <0.001 2,200) 10.64) 9.63) 6.15) 3.68)
PFNA 1.40(1.10, 1.80) 1.40(1.10, 1.80) <0.001 CD3þ T cells 1,356 7.43 5.89 3.12 1.30
(1,085, (3.80, (1.46, (0.20, (-1.25,
Numbers (%) or median (interquartile range) are presented. BMI, Body mass
1,645) 11.19) 10.51) 6.13) 3.92)
index (only available for the 2005–2006 survey); GED, General education
CD3þCD4þT-helper 904 8.28 7.42 3.94 2.04
development; HSD, High school diploma. PFAS, Perfluoroalkyl substance. cells (714, (4.28, (2.78, (0.60, (-0.84,
PFHxS, Perfluorohexane sulfonate; PFNA, Perfluorononanoic acid; PFOA, Per­ 1105) 12.44) 12.26) 7.40) 5.01)
fluorooctanoic acid; PFOS, Perfluorooctane sulfonate. CD3þCD8þT- 401 5.51 3.99 1.62 − 0.88
a
Likelihood ratio test p-value from unadjusted ordinal mixed models (for cytotoxic cells (293, (0.52, (-2.40, (-2.31, (-3.97,
sociodemographic covariates), or linear mixed models adjusted for gender, age, 534) 10.75) 10.80) 5.71) 2.32)
smoking, month of sampling, alcohol intake, and educational level (for ln CD3þCD4þCD8þDP 16 (10, 6.03 12.21 − 0.23 1.06
(PFAS)). T cells 25) (-1.21, (2.47, (-5.68, (-4.01,
13.79) 22.89) 5.55) 6.40)
CD3¡ 184 8.62 4.94 5.40 0.31
PFOS and PFNA in 2010) (Figure S1). CD16þCD56þNK (129, (3.45, (-0.75, (1.35, (-3.34,
Most median cell counts were slightly lower in the later survey cells 260) 14.04) 10.96) 9.60) 4.10)
CD3¡ CD19þB cells 238 7.26 3.32 3.89 2.26
sample, though this was not the case for eosinophils (Table 2). Median
(170, (2.08, (-2.82, (-0.64, (-1.79,
percentages of the different immune cells were mostly very similar be­ 318) 12.70) 9.86) 8.63) 6.47)
tween the two surveys (Table 3). CD4þ/CD8þratio 2.35 2.63 3.29 2.28 2.94
(1.71, (-2.19, (-2.53, (-1.78, (-0.31,
3.02) 7.68) 9.46) 6.52) 6.30)
3.2. Serum PFASs and WBC count
Immune cell count > 0 in 100% of samples except for monocytes (99.9% and
For total WBC count in the 2005–2006 survey, positive (PFHxS) and 99.6% in 2005–2006 and 2010, respectively) and eosinophils (91.9% and
inverse (rest of the contaminants) associations were found, reaching 93.2%). PFASs and counts of immune cells were log transformed (ln) for ana­
statistical significance for PFHxS and PFOS (difference: 0.55% and lyses. Likelihood ratio test p-value < 0.001 for the comparison between the two
time points for all outcomes except eosinophils (linear mixed models). Cell
− 0.55% per PFHxS and PFOS IQR increment, corresponding to 39 and
counts are expressed as cells/μL. Results are expressed as the difference in the
− 39 cells/μl when referring to the median population count). WBC as­
outcomes associated with IQR increments in PFAS levels in 2005–2006. All
sociations showed wide confidence intervals in the smaller follow-up models were adjusted for gender, age, smoking, month of sampling, alcohol
population (Tables 2 and 3 and S1 and Fig. 1 and S2). intake, and educational level.
% diff., % difference; CI, Confidence interval; DP, Double positive; IQR, Inter­
3.3. Serum PFASs and myeloid lineage quartile range; NK, Natural killer; PFASs, Perfluoroalkyl substances; PFHxS,
Perfluorohexane sulfonate; PFNA, Perfluorononanoic acid; PFOA, Per­
There was some evidence of an association between PFOA, PFOS, fluorooctanoic acid; PFOS, Perfluorooctane sulfonate; WBCs, White blood cells.
PFNA, and neutrophil count (difference range: − 1.56% to − 0.77% per
PFAS IQR increment [–69 to –34 cells/μl]) and between the four PFASs
and neutrophil percentage (− 0.65 to − 0.36 per PFAS IQR increment) in

4
M.-J. Lopez-Espinosa et al. Environment International 156 (2021) 106599

Table 3 2010 survey. Finally, eosinophils were associated with PFHxS in the first
Percentages of WBCs and lymphocyte subsets, and association between PFASs survey (difference: − 0.96% and − 0.04 for count and percentage, per
and percentages of these cells amongst the population in 2005–2006 (n = PFHxS IQR increment, respectively) and PFNA in both surveys, but the
42,782) and in 2010 (n = 526), Mid-Ohio Valley, USA. direction of the latter association changed between the two time points
Cell PFHxS PFOA PFOS PFNA (difference: − 0.76% and 6.37% for count and − 0.03 and 0.13 for per­
percentage centage, per PFNA IQR increment) (Tables 2–3, and Fig. 1 and S2).
Outcome Median diff. diff. diff. diff. Given the smaller contribution of monocytes and eosinophils to the WBC
(IQR) (95% (95% (95% (95% count, such changes were almost negligible (range: 1–6 cells/μl) when
CI) CI) CI) CI) expressed in absolute terms (Table S1).
Neutrophils 62 (57, 67)a − 0.36 − 0.41 − 0.65 − 0.37
2005–2006 (-0.46, (-0.52, (-0.74, (-0.45,
− 0.26) − 0.30) − 0.55) − 0.29) 3.4. Serum PFASs and lymphoid lineage
Neutrophils 2010 63 (57, 68)a − 1.74 − 1.41 − 0.85 − 0.58
(-2.54, (-2.38, (-1.63, (-1.31, For total lymphocytes, positive associations were clearly evident for
− 0.94) − 0.44) − 0.07) 0.15)
PFHxS, PFOA, and PFOS. These associations were stronger for absolute
Monocytes 6.0 (5.0, 0.02 0.04 0.04 0.00
2005–2006 7.0)a (0.00, (0.02, (0.02, (-0.02, numbers compared to percentages and with a larger effect size in the
0.05) 0.07) 0.06) 0.02) 2010 subset compared to the 2005–2006 population. Specifically, the
Monocytes 2010 5.0 (4.0, 0.02 0.16 0.03 0.04 relative (percent) differences in the counts of lymphocytes for an IQR
7.0)a (-0.2, (-0.08, (-0.15, (-0.11, contrast in PFHxS was 2.00% in the large population and 7.33% in the
0.24) 0.39) 0.20) 0.20)
Eosinophils 2.0 (1.0, − 0.04 0.00 − 0.01 − 0.03
follow-up, 1.12% and 5.50% for PFOA, and 1.95% and 3.39% for PFOS
2005–2006 3.0)a (-0.06, (-0.02, (-0.03, (-0.04, (Table 2). Concerning absolute counts, these estimates represented
− 0.02) 0.02) 0.01) − 0.01) changes ranging from 22 to 132 cells/μl (Table S1). Respective data for
Eosinophils 2010 2.0 (1.0, − 0.04 0.05 0.06 0.13 the difference increase in lymphocyte percentage were 0.37 and 1.77
3.0)a (-0.23, (-0.21, (-0.11, (0.01,
(PFHxS), 0.36 and 1.24 (PFOA), and 0.61 and 0.77 (PFOS) (Table 3).
0.15) 0.32) 0.22) 0.26)
Lymphocytes 29 (24, 34)a 0.37 0.36 0.61 0.39 Fig. 1 and S2 show these graphically across PFAS deciles (population in
2005–2006 (0.28, (0.26, (0.53, (0.32, 2005–2006) and tertiles (population in 2010) with, in general, clear
0.46) 0.45) 0.69) 0.46) monotonic increases for these three contaminants and lymphocytes. A
Lymphocytes 2010 29 (24, 34)a 1.77 1.24 0.77 0.43 monotonic association was also found between PFNA and lymphocyte
(1.05, (0.36, (0.13, (-0.23,
2.50) 2.12) 1.42) 1.08)
cells (difference for count and percentage: 1.47% [29 cells/μl] and 0.39
CD3þT cells 75 (70, 79)b 0.02 0.27 − 0.20 0.00 per PFNA IQR increment) in the 2005–2006 population but not in that of
(-0.70, (-0.54, (-0.79, (-0.52, 2010 (Tables 2–3 and Fig. 1 and S2).
0.74) 1.07) 0.39) 0.52) The above-mentioned statistically significant changes in absolute
CD3þCD4þT-helper 50 (45, 55)b 0.45 0.88 0.24 0.40
lymphocyte count led to changes in the numbers of lymphocyte subsets.
cells (-0.38, (-0.05, (-0.45, (-0.15,
1.28) 1.82) 0.93) 0.95) Specifically, we found associations between most of the types of
CD3þCD8þT- 22 (17, 27)b − 0.44 − 0.36 − 0.51 − 0.42 numbers of lymphocyte subsets and PFHxS concentrations, with differ­
cytotoxic cells (-1.29, (-1.40, (-1.26, (-1.01, ences ranging from 5.51% in CD3+ CD8+ T-cytotoxic cells to 8.62% in
0.41) 0.68) 0.24) 0.17) CD3− CD16+ CD56+ NK cells per PFHxS IQR increment (Table 2).
CD3þCD4þCD8þDP 0.9 (0.6, − 0.03 0.15 − 0.05 0.02
T cells 1.3)b (-0.12, (-0.02, (-0.13, (-0.07,
Fig. 2 shows clear monotonic increases across PFHxS tertiles for all B,
0.07) 0.33) 0.03) 0.11) NK, and T cell numbers except the CD3+ CD4+ CD8+ DP subset. For
CD3¡ 10 (7.3, 0.16 0.04 0.27 − 0.04 other PFASs the associations were more limited. For PFOA, associations
CD16þCD56þNK 14)b (-0.37, (-0.62, (-0.12, (-0.44, with the numbers of the above-mentioned lymphocytes were also
cells 0.70) 0.71) 0.66) 0.37)
evident for CD3+ T cells (5.89%) and CD3+ CD4+ T-helper cells
CD3¡ CD19þB cells 13 (10, 17)b − 0.18 − 0.33 − 0.04 0.06
(-0.77, (-0.93, (-0.55, (-0.39, (7.42%), and more strongly for CD3+ CD4+ CD8+ DP T cells (12.21%).
0.42) 0.28) 0.46) 0.51) For PFOS, again there were associations with both CD3+ and CD3+
CD4+ T cells, and CD3− CD16+ CD56+ NK cells (3.12%, 3.94%, and
Immune cell count > 0 in 100% of samples except for monocytes (99.9% and
99.6% in 2005–2006 and 2010, respectively) and eosinophils (91.9% and
5.40%, respectively) (Table 2). The magnitudes of the associations
93.2%). PFASs were log transformed (ln) for analyses. Likelihood ratio test p- increased more clearly monotonically across tertiles for both CD3+ T
value < 0.001 for the comparison between both time points for all outcomes cells and CD3+ CD4+ T cells in relation to PFOA, and CD3− CD16+
except lymphocytes and eosinophils (linear mixed models). Results are CD56+ NK cells in relation to PFOS (Fig. 2). PFNA was not associated
expressed as the difference in the outcomes associated with IQR increments in with any subtype of lymphocyte (Table 2 and Fig. 2).
PFAS levels in 2005–2006. All models were adjusted for gender, age, smoking, Regarding percentage data, associations were mostly null in all cases,
month of sampling, alcohol intake, and educational level. meaning that changes in actual cell numbers strongly resembled those
CI, Confidence interval; diff., difference; DP, Double positive; IQR, Interquartile seen for total lymphocytes, but now distributed accordingly across the
range; NK, Natural killer; PFASs, Perfluoroalkyl substances; PFHxS, Per­ different phenotypes (Table 3 and Figure S3).
fluorohexane sulfonate; PFNA, Perfluorononanoic acid; PFOA, Per­
fluorooctanoic acid; PFOS, Perfluorooctane sulfonate.
a 3.5. Sensitivity analyses
Percentage of total WBCs.
b
Percentage of total lymphocytes.
Various models including other variables which might have
confounded the results are set out in detail in the Supplementary ma­
the first survey. The effect was notably larger for the percentage of
terial (Figures S4–S9). Firstly, modest rather than major changes in the
neutrophils in relation to PFHxS, PFOA, and PFOS in the follow-up
associations resulted after adding BMI, eGFR, or anti-inflammatory
population (difference: − 1.74, − 1.41, and − 0.85, per PFAS IQR incre­
drugs, or excluding self-reported cancer or autoimmune diseases. Sec­
ment, respectively). We also found some evidence of a positive associ­
ondly, in models containing all four PFASs, the regression coefficients
ation between PFHxS, PFOA, and monocyte count (difference: 1.16%
fell or were even close to null in some cases and their CIs were wider.
and 0.61% per PFAS IQR increment, respectively) and between PFHxS,
Specifically, for the myeloid lineage bigger changes were shown in the
PFOA, PFOS, and monocyte percentage (difference range: 0.02 to 0.04
2005–2006 survey compared to that of 2010. Regarding lymphocytes,
per PFAS IQR increment, respectively) in the 2005–2006 but not in the
the association was somewhat smaller for PFHxS versus count and null

5
M.-J. Lopez-Espinosa et al. Environment International 156 (2021) 106599

Fig. 1. Adjusted WBC counts (mean and 95% confidence interval) by PFAS deciles (2005–2006, n ¼ 42,782) or tertiles (2010, n ¼ 526), Mid-Ohio Valley,
USA. Statistically significant trends at p < 0.05 and p < 0.01 are denoted by one and two asterisks, respectively. All models were adjusted for gender, age, smoking,
month of sampling, alcohol intake, and educational level. PFAS, Perfluoroalkyl substance; PFHxS, Perfluorohexane sulfonate; PFNA, Perfluorononanoic acid; PFOA,
Perfluorooctanoic acid; PFOS, Perfluorooctane sulfonate, WBCs, White blood cells.

for percentage in the 2005–2006 survey. For PFOA, the associations No clear association was found between total WBC count and PFAS
were smaller, reaching the null in 2005-2005 for count and losing sta­ levels, statistical significance only being reached for PFHxS and PFOS in
tistical significance for count and percentage in 2010. For PFOS and both the 2005–2006 survey. The effects were minimal, and the direction of
count and percentage the associations were smaller in 2005–2006 and the association was not consistent with PFHxS, showing a positive as­
disappeared in 2010. For numbers of lymphocyte subtypes including the sociation and the others showing negative trends. Two studies
four PFASs simultaneously, the associations were, in general, a little measuring serum PFOA and PFOS in workers at 3M (n = 53) and DuPont
smaller for PFHxS and PFOA but completely disappeared for PFOS. (n = 518) facilities located in Italy, Alabama (USA), and Belgium did not
Thirdly, multivariate analysis amongst the full follow-up population, i. report any significant associations between these contaminants and total
e., population reporting (or not) infections in 2010 (n = 736) also sug­ WBC count (range of PFOA and PFOS means were much higher than in
gested associations between PFHxS, PFOA, and PFOS in relation to im­ the present study: 0.07–19.7 and 0.13–1.40 μg/mL, respectively) (Costa
mune cell counts, but with generally lower % differences and some with et al., 2009; Olsen et al., 2003). Diverging results have been reported in
CIs including the null. Finally, although some p-values of the interaction non-occupational studies. Specifically, in a study on a population (n =
with sex were < 0.05, especially for the WBC types, no clear evidence of 371) with longstanding environmental PFOA exposure (median: 354
a differential effect between males and females was found on consid­ ng/mL) in the same area as the present study, marginally significant
ering the two surveys. positive associations with WBC count were found (Emmett et al., 2006).
A cross-sectional study in pregnant women (n = 189) in Greenland
4. Discussion (Knudsen et al., 2018) reported a significant negative association be­
tween the sum of 17 PFASs (ΣPFAS median: 18.5 ng/mL) and WBC
We determined peripheral WBC counts in two surveys (2005–2006 count. However, levels of 4 PFASs in maternal and child samples at 18
and 2010) and lymphocyte subtypes in 2010 to characterize potential months and 5 years of age (ΣPFAS geometric mean range: 0.47–0.73 ng/
health effects related to the immune system associated with PFAS con­ mL) were not associated with WBC count at 5 years of age in a cohort (n
centrations. Our community-based sample had a wide range of PFOA = 56) of Faroese children (Oulhote et al., 2017).
serum concentrations (IQR: 13.2–69.2 ng/mL; median: 26.9 ng/mL in Examination of cell differentials resulted in mixed associations for
2005–2006), and background levels of PFHxS, PFOS, and PFNA (me­ the myeloid lineage. Neutrophils showed slightly decreased trends
dians: 2.9, 19.7, and 1.4 ng/mL in 2005–2006, respectively). The trends associated with PFASs in the first survey and less clearly in the second
with PFASs for WBCs were slight, with small percentage differences survey, where the association was only with percentages of this cell type.
across the IQRs. For most PFASs there were negative slopes for neutro­ In line with our results, negative associations were reported between the
phils, with larger positive slopes for lymphocytes. Our results suggest a sum of 17 PFASs and numbers of neutrophils in the above-mentioned
consistent positive association between serum PFHxS and absolute pregnant women from Greenland (Knudsen et al., 2018). Conversely,
lymphocyte count and weaker evidence of an association for percentage other studies did not find such an association (Dong et al., 2013; Emmett
of total lymphocytes. For PFHxS and absolute counts of lymphocyte et al., 2006; Oulhote et al., 2017). Regarding other myeloid cells, we
subtypes, significant and positive associations were found for T, B, and found positive associations between PFHxS, PFOA, and PFOS and
NK cell counts, which likely reflect the changes in the absolute monocytes in the first survey, while no consistent pattern of an associ­
lymphocyte count. A less consistent association was also shown for both ation was found for monocytes in the second survey or for eosinophils. In
PFOA and PFOS and total and subtypes of lymphocytes. the above-mentioned study on the Mid-Ohio Valley, significant positive

6
M.-J. Lopez-Espinosa et al. Environment International 156 (2021) 106599

some cases. Since counts of lymphocyte subtypes are not independent of


the absolute lymphocyte count and there were only significant changes
in counts but not in percentages of the lymphocyte subsets, the observed
changes likely reflect changes in absolute lymphocyte counts rather than
any specific effect on lymphocyte subsets themselves. Something similar
has been reported in smoker’s leukocytosis, a well-documented effect of
active smoking (Pedersen et al., 2019; Sopori, 2002) where the effect of
smoking raises WBCs without greatly changing subset percentages of the
different cell types (Park et al., 1992). With regard to PFASs, only two
epidemiological studies, both focused on child populations, have been
conducted on peripheral blood lymphocytes and PFASs, and no signifi­
cant associations were reported (Abraham et al., 2020; Oulhote et al.,
2017). On a different note, animal experiments have reported altered
lymphocyte populations following exposure to PFASs, although effects
seem species- or even strain-dependent (DeWitt et al., 2009). Increased
CD3+ CD4+ T-helper cells and decreased CD3+ CD8+ T-cytotoxic cells
in spleens and increased CD3+ CD8+ T–cytotoxic cells in thymus were
reported in male ICR mice exposed to PFOA for 21 days (Son et al.,
2009). Reduced B and T cells were also reported, particularly CD4+
CD8+ thymocytes in male C57BL/6 mice treated with PFOA for 7 or 10
days (Yang et al., 2001). In contrast, our finding of a positive association
between PFAS exposure and CD3− CD16+ CD56+ NK cells might be
more consistent with the increase in NK cell activity reported in B6C3F1
male mice exposed to PFOS (Peden-Adams et al., 2008).
Fig. 2. Adjusted lymphocyte subtype counts (mean and 95% confidence In the present study, despite the evidence of an association with
interval) by PFAS tertiles (n ¼ 526), Mid-Ohio Valley, USA (2010). Sta­
increased total lymphocyte cells, the absolute magnitudes of difference
tistically significant trends at p < 0.05 and p < 0.01 are denoted by one and two
in cell numbers were small (difference range: 1.12–7.33% per PFAS IQR
asterisks, respectively. All models were adjusted for gender, age, smoking,
month of sampling, alcohol intake, and educational level. DP, Double positive; increment, which when compared to the population median represented
NK, Natural killer; PFAS, Perfluoroalkyl substance; PFHxS, Perfluorohexane at most 132 [95% CI: 74, 191] cells/μL for PFHxS in 2010) and the
sulfonate; PFNA, Perfluorononanoic acid; PFOA, Perfluorooctanoic acid; PFOS, percentages of total WBCs that these cells represented changed very
Perfluorooctane sulfonate. little (difference range: 0.36–1.77 per PFAS IQR increment). Therefore,
lymphocyte changes in the present study cannot be considered clinically
meaningful. The same is true for the decreases observed in absolute
associations with monocyte count but not percentage were found neutrophil count: though statistically significant for some PFASs, espe­
(Emmett et al., 2006). Interestingly, none of the above-mentioned im­ cially for the 2005–2006 data, these were quite small and would not be
mune cell types measured in children aged 5 years were associated with clinically important in themselves. Further studies would be needed to
prenatal, 18-month or 5-year concentrations of PFASs in Faroese chil­ determine whether these effects could be meaningful in specific clinical
dren (Oulhote et al., 2017). Cross-sectional studies reported conflicting contexts, such as a serious bacterial infection.
results for eosinophil counts, with one study finding a positive associa­ Overall, these findings, along with previous findings of suppressed
tion with PFOA (median in cases [n = 231] and controls [n = 225]: 1.2 antibody response (Abraham et al., 2020; Grandjean et al., 2017, 2012;
and 0.5 ng/mL, respectively) and PFOS (33.9 and 28.9 ng/mL) among Kielsen et al., 2016; Looker et al., 2014), suggest that the effect of PFASs
asthmatic compared to nonasthmatic Taiwanese teenagers (Dong et al., on immune function, if there is one, is not mediated solely through
2013). However, another study reported null results for 6 PFAS conge­ altered lymphocyte numbers but by changes in cell signaling or function
ners (median ΣPFASs for cases [n = 118] and controls [n = 169]: 7.3 and (as measured by antibody response).
5.7 ng/mL) among New Yorkers aged 16–17 years exposed to high levels Previous studies in the population considered here have suggested
of PFASs, among other contaminants, in the World Trade Center disaster that impaired kidney function may lead to decreased net PFOA excretion
who were matched to a control group (Gaylord et al., 2019). via the kidneys and, accordingly, increased serum levels of PFOA
Results were clearer for total lymphocytes, showing statistically (Watkins et al., 2013). Thus, confounding by kidney function may
significant positive and generally monotonic associations with all PFAS plausibly occur due to the fact that inflammation or ill health, which can
levels except PFNA in the two surveys. Specifically, PFHxS presented the be associated with renal impairment, may also be reflected in the
strongest effect for a small absolute contrast in levels (i.e., for an IQR elevated lymphocyte count. Since eGFR reflects the filtering capacity of
contrast of only 2.8 ng/mL it showed the biggest increase with both the kidney (Dhingra et al., 2017; Watkins et al., 2013) and was available
count and percentage of lymphocytes). For PFOA and PFOS, there was for the study population, we repeated the regression analysis including
also an association, but less clear. With regard to the previous epide­ eGFR in the models, with no evidence of eGFR confounding the asso­
miological studies measuring lymphocytes, none have reported PFAS ciation between PFASs and immune cell outcomes. Estimates in the main
exposure as being associated with alterations in this particular immune analysis did not differ markedly when adding other possible con­
cell type during childhood (Abraham et al., 2020; Oulhote et al., 2017) founders such as 2005–2006 BMI or self-reported anti-inflammatory
or adulthood (Emmett et al., 2006; Knudsen et al., 2018). drug therapy (only available for the follow-up study). The main differ­
To better characterize the lymphocytosis and help determine ences were found for the inclusion of the four PFASs at the same time
whether there was any evidence of altered immunoregulation, immu­ since CIs were wider, and the estimates fell when including other PFASs
nophenotyping of peripheral blood was conducted in the 2010 survey to or were even close to null in some cases. The exclusion of people who
enumerate the major lymphocyte populations. For lymphocyte sub­ reported autoimmune disease (both surveys) or cancer (first survey)
types, PFHxS concentrations were positively and monotonically associ­ hardly modified the estimated associations. Finally, gender-specific
ated with absolute counts of T, B, and NK cells. In addition, higher PFOA differences in the development of reproductive organs and physiology
serum levels were positively associated with some T cell subsets and may result in gender-specific responses to a given toxicant (Bach et al.,
PFOS with some T cell subsets and NK cells, with monotonicity shown in 2015). For PFASs, two studies have indicated a higher vulnerability

7
M.-J. Lopez-Espinosa et al. Environment International 156 (2021) 106599

among asthmatic boys compared to girls (Qin et al., 2017; Zhu et al., CRediT authorship contribution statement
2016). Mixed results were reported in another study with girls or boys at
higher risk depending on the outcome (asthma, atopic dermatitis, and Maria-Jose Lopez-Espinosa: Conceptualization, Data curation,
rhinitis) and the class of PFAS studied (Kvalem et al., 2020). However, Formal analysis, Investigation, Methodology, Software, Validation,
although we found some significant gender interactions indicating that Visualization, Writing - original draft, Writing - review & editing.
higher PFAS exposure was associated with higher counts of types of Christian Carrizosa: Data curation, Formal analysis, Investigation,
WBCs in females, and lower counts in males, such a pattern was not Methodology, Software, Visualization, Writing - original draft, Writing -
consistent. review & editing. Michael I. Luster: Conceptualization, Investigation,
Our study has several limitations. First, information on residential Methodology, Resources, Validation, Validation, Writing - review &
histories, infections, and other potential confounders was self-reported, editing. Joseph B. Margolick: Investigation, Methodology, Resources,
and participants were only asked about the presence or absence (not Visualization, Writing - original draft, Writing - review & editing. Olga
severity) of specific infections. Nevertheless, restricting our analysis to Costa: Data curation, Formal analysis, Investigation, Methodology,
those who reported no infections in the 2010 survey should have Software, Writing - review & editing. Giovanni S. Leonardi: Concep­
reduced potential bias due to infection reports. Second, information on tualization, Funding acquisition, Investigation, Methodology, Valida­
infections was only available for the follow-up population and BMI only tion, Writing - review & editing. Tony Fletcher: Conceptualization,
for the first survey. Nevertheless, the results for participants who did not Data curation, Formal analysis, Funding acquisition, Investigation,
report infection in 2010 (n = 526) are somewhat stronger than those for Methodology, Project administration, Resources, Supervision, Valida­
the full population in 2010 (n = 736) or for the 2005–2006 cohort. In tion, Visualization, Writing - original draft, Writing - review & editing.
addition, sensitivity analyses including 2005–2006 BMI did not sub­
stantially change the results in either of the two surveys, thus ruling out
Declaration of Competing Interest
any role of BMI as a confounder. Furthermore, in this population PFOA
exposure was largely driven by residential water district and consump­
The authors declare that they have no known competing financial
tion, and less correlated with individual self-reported characteristics and
interests or personal relationships that could have appeared to influence
behaviors (Steenland et al., 2009). Third, given the high-exposure na­
the work reported in this paper.
ture of our population for PFOA, the results for this contaminant may not
be generalizable to other populations. Fourth, the immune system has
many components but we studied only the absolute counts and per­ Acknowledgments
centages of the main types of immune cells, with no assessment of the
function of these cells. However, it is pertinent to highlight that, in a The authors wish to thank the participants for their contributions to
previous study of part of the follow-up population of the present study, this study. We are also grateful to the Centers for Disease Control and
increased PFOA concentrations were associated with a lower antibody Prevention (CDC) laboratory for analyzing the PFASs in serum samples.
response to A/H3N2 influenza vaccine (Looker et al., 2014). Fifth, due to This project was funded by the C8 Class Action Settlement Agree­
the high number of statistical analyses, there is the possibility of po­ ment (Circuit Court of Wood County, WV) between DuPont and plain­
tential false positive statistically significant associations (i.e., type I er­ tiffs, which resulted from releases of perfluorooctanoate (PFOA, or C8)
rors). The estimates for the coefficients and their confidence intervals into drinking water. It was one of the C8 Science Panel Studies under­
should be taken as a global picture of the pattern of the relations be­ taken by the Court-approved C8 Science Panel, of which Dr Tony
tween the variables involved in the study. Finally, humans are poten­ Fletcher was a member, established under the same Settlement Agree­
tially exposed to a large variety of other types of contaminants, and ment. Dr Fletcher is a consortium partner of the European Union’s Ho­
further study of the relation between exposures to multiple chemicals rizon 2020 research and innovation program under grant agreement
with immunotoxic capability is warranted. 733032 HBM4EU (www.HBM4EU.eu); part-funded by the National
The major strengths of this study are the large sample size, assess­ Institute for Health Research (NIHR) Health Protection Research Unit in
ments of PFAS levels and some immune cells at two time points with an Environmental Exposures and Health, a partnership between Public
interval of approximately five years, and the inclusion of individual Health England, the Health and Safety Executive, and the University of
potential confounders. In addition, it is believed that these participants Leicester; and part funded with a PFAS research grant from CORIS/
are representative, at least for the first survey, given the high partici­ REGIONE VENETO (Italy). The views expressed are those of the author
pation rates in the C8 Health Project (~85% in the range of age of the (s) and not necessarily those of the NIHR, Public Health England, the
present study) (Frisbee et al., 2009) of those adults who drank Health and Safety Executive or the Department of Health and Social
contaminated water in the Mid-Ohio Valley, and this diminishes concern Care. MJ Lopez-Espinosa holds grants from the Spanish Carlos III Health
about potential selection biases. Thirdly, despite an increasing body of Institute (Miguel Servet-FSE: MSII16/00051 and FIS-FEDER: PI14/
literature on the possible immunotoxic effects of PFASs in humans and 00891 and PI17/00663), Alicia Koplowitz Foundation 2017, and Min­
specific animal models demonstrating specific lymphocyte changes, to istry of Education, Culture and Sports (Jose Castillejo Grant: CAS17/
our knowledge studies involving human immunophenotypes are scarce 00052).
and there is therefore a gap in the literature on this topic.
In summary, the strongest association was observed between abso­ Appendix A. Supplementary material
lute lymphocyte count and PFHxS. If causal, our results suggest that
PFHxS may be more potent compared to the other PFASs, since a Supplementary data to this article can be found online at https://doi.
contrast of only 2.8 ng/ml was associated with as much as a 7% increase org/10.1016/j.envint.2021.106599.
in absolute lymphocyte count in the 2010 survey. Weaker evidence of an
association was also shown for PFHxS and percentage of lymphocytes. References
For PFOA and PFOS, the association was less clear. For all PFASs, the
magnitudes of the differences in absolute lymphocyte count were small. Abraham, K., Mielke, H., Fromme, H., Völkel, W., Menzel, J., Peiser, M., Zepp, F.,
Willich, S.N., Weikert, C., 2020. Internal exposure to perfluoroalkyl substances
A similar pattern was found for lymphocyte cell subtypes with, again, (PFASs) and biological markers in 101 healthy 1-year-old children: associations
stronger evidence for PFHxS. Nevertheless, the changes observed in between levels of perfluorooctanoic acid (PFOA) and vaccine response. Arch.
absolute counts of lymphocyte subsets reflected the change in total Toxicol. 94, 2131–2147. https://doi.org/10.1007/s00204-020-02715-4.
Agency for Toxic Substances and Disease Registry [ATSDR], 2018. Toxicological profile
lymphocyte count rather than any specific effect on the subsets them­ for perfluoroalkyls (Draft for public comment). Atlanta, GA: US Department of
selves, since no association was reported for percentage of the subtypes. Health and Human Services, Public Health Service.

8
M.-J. Lopez-Espinosa et al. Environment International 156 (2021) 106599

Aziz, N., Margolick, J.B., Detels, R., Rinaldo, C.R., Phair, J., Jamieson, B.D., Butch, A.W., Grandjean, P., Heilmann, C., Weihe, P., Nielsen, F., Mogensen, U.B., Budtz-Jørgensen, E.,
2013. Value of a quality assessment program in optimizing cryopreservation of 2017. Serum vaccine antibody concentrations in adolescents exposed to
peripheral blood mononuclear cells in a multicenter study. Clin. Vaccine Immunol. perfluorinated compounds. Environ. Health Perspect. 125, 077018 https://doi.org/
20, 590–595. https://doi.org/10.1128/CVI.00693-12. 10.1289/EHP275.
Bach, C.C., Bech, B.H., Brix, N., Nohr, E.A., Bonde, J.P.E., Henriksen, T.B., 2015. Haase, H., Fahlenkamp, A., Schettgen, T., Esser, A., Gube, M., Ziegler, P., Kraus, T.,
Perfluoroalkyl and polyfluoroalkyl substances and human fetal growth: A systematic Rink, L., 2016. Immunotoxicity monitoring in a population exposedto
review. Crit. Rev. Toxicol. 45, 53–67. https://doi.org/10.3109/ polychlorinated biphenyls. Int. J. Environ. Res. Public Health 13, 1–13. https://doi.
10408444.2014.952400. org/10.3390/ijerph13030295.
Ballesteros, V., Costa, O., Iñiguez, C., Fletcher, T., Ballester, F., Lopez-Espinosa, M.J., International Pollutants Elimination Network, 2019. PFAS pollution across the Middle
2017. Exposure to perfluoroalkyl substances and thyroid function in pregnant East and Asia [WWW Document]. accessed 6.8.20. https://www.documentcloud.
women and children: A systematic review of epidemiologic studies. Environ. Int. 99, org/documents/5980916-Pfas-Pollution-Across-the-Middle-East-and-Asia.html.
15–28. https://doi.org/10.1016/j.envint.2016.10.015. Kato, K., Basden, B.J., Needham, L.L., Calafat, A.M., 2011. Improved selectivity for the
Braun, J.M., 2017. Early-life exposure to EDCs: Role in childhood obesity and analysis of maternal serum and cord serum for polyfluoroalkyl chemicals.
neurodevelopment. Nat. Rev. Endocrinol. 13, 161–173. https://doi.org/10.1038/ J. Chromatogr. A 1218, 2133–2137. https://doi.org/10.1016/j.
nrendo.2016.186. chroma.2010.10.051.
Cardenas, A., Hauser, R., Gold, D.R., Kleinman, K.P., Hivert, M.F., Fleisch, A.F., Lin, P.I. Kielsen, K., Shamim, Z., Ryder, L.P., Nielsen, F., Grandjean, P., Budtz-Jørgensen, E.,
D., Calafat, A.M., Webster, T.F., Horton, E.S., Oken, E., 2018. Association of Heilmann, C., 2016. Antibody response to booster vaccination with tetanus and
Perfluoroalkyl and Polyfluoroalkyl Substances With Adiposity. JAMA Netw. open 1, diphtheria in adults exposed to perfluorinated alkylates. J. Immunotoxicol. 13,
e181493. https://doi.org/10.1001/jamanetworkopen.2018.1493. 270–273. https://doi.org/10.3109/1547691X.2015.1067259.
Castilho, J.L., Shepherd, B.E., Koethe, J., Turner, M., Bebawy, S., Logan, J., Rogers, W.B., Knudsen, A.K.S., Long, M., Pedersen, H.S., Bonefeld-Jørgensen, E.C., 2018. Persistent
Raffanti, S., Sterling, T.R., 2016. CD4+/CD8+ratio, age, and risk of serious organic pollutants and haematological markers in Greenlandic pregnant women: the
noncommunicable diseases in HIV-infected adults on antiretroviral therapy. AIDS ACCEPT sub-study. Int. J. Circumpolar Health 77, 1456303. https://doi.org/
30, 899–908. https://doi.org/10.1097/QAD.0000000000001005. 10.1080/22423982.2018.1456303.
Chen, Q., Huang, R., Hua, L., Guo, Y., Huang, L., Zhao, Y., Wang, X., Zhang, J., 2018. Kvalem, H.E., Nygaard, U.C., Lødrup Carlsen, K.C., Carlsen, K.H., Haug, L.S., Granum, B.,
Prenatal exposure to perfluoroalkyl and polyfluoroalkyl substances and childhood 2020. Perfluoroalkyl substances, airways infections, allergy and asthma related
atopic dermatitis: A prospective birth cohort study. Environ. Heal. 17, 8. https://doi. health outcomes – implications of gender, exposure period and study design.
org/10.1186/s12940-018-0352-7. Environ. Int. 134, 105259 https://doi.org/10.1016/j.envint.2019.105259.
Costa, G., Sartori, S., Consonni, D., 2009. Thirty years of medical surveillance in Lau, C., Anitole, K., Hodes, C., Lai, D., Pfahles-Hutchens, A., Seed, J., 2007.
perfluooctanoic acid production workers. J. Occup. Environ. Med. 51, 364–372. Perfluoroalkyl acids: A review of monitoring and toxicological findings. Toxicol. Sci.
https://doi.org/10.1097/JOM.0b013e3181965d80. 99, 366–394. https://doi.org/10.1093/toxsci/kfm128.
Davis, A.S., Viera, A.J., Mead, M.D., 2014. Leukemia: An overview for primary care. Am. Leonardi, G.S., Houthuijs, D., Steerenberg, P.A., Fletcher, T., Armstrong, B., Antova, T.,
Fam. Physician 89, 731–738. Lochman, I., Lochmanova, A., Rudnai, P., Erdei, E., Musial, J., Jazwiec-Kanyion, B.,
DeWitt, J.C., Shnyra, A., Badr, M.Z., Loveless, S.E., Hoban, D., Frame, S.R., Cunard, R., Niciu, E.M., Durbaca, S., Fabianova, E., Koppova, K., Lebret, E., Brunekreef, B., Van
Anderson, S.E., Meade, B.J., Peden-Adams, M.M., Luebke, R.W., Luster, M.I., 2009. Loveren, H., 2000. Immune biomarkers in relation to exposure to particulate matter:
Immunotoxicity of perfluorooctanoic acid and perfluorooctane sulfonate and the role A cross-sectional survey in 17 cities of Central Europe. Inhal. Toxicol. 12, 1–14.
of peroxisome proliferator-activated receptor alpha. Crit. Rev. Toxicol. https://doi. https://doi.org/10.1080/089583700750019486.
org/10.1080/10408440802209804. Levey, A.S., Coresh, J., Greene, T., Stevens, L.A., Zhang, Y., Hendriksen, S., Kusek, J.W.,
Dhingra, R., Winquist, A., Darrow, L.A., Klein, M., Steenland, K., 2017. A study of reverse Van Lente, F., 2006. Using standardized serum creatinine values in the Modification
causation: Examining the associations of perfluorooctanoic acid serum levels with of Diet in Renal Disease study equation for estimating glomerular filtration rate.
two outcomes. Environ. Health Perspect. 125, 416–421. https://doi.org/10.1289/ Ann. Intern. Med. 145, 247–254. https://doi.org/10.7326/0003-4819-145-4-
EHP273. 200608150-00004.
Dong, G.H., Tung, K.Y., Tsai, C.H., Liu, M.M., Wang, D., Liu, W., Jin, Y.H., Hsieh, W.S., Long, J.S., Ervin, L.H., 2000. Using heteroscedasticity consistent standard errors in the
Lee, Y.L., Chen, P.C., 2013. Serum polyfluoroalkyl concentrations, asthma outcomes, linear regression model. Am. Stat. 54, 217–224. https://doi.org/10.1080/
and immunological markers in a case-control study of Taiwanese children. Environ. 00031305.2000.10474549.
Health Perspect. 121, 507–513. https://doi.org/10.1289/ehp.1205351. Looker, C., Luster, M.I., Calafat, A.M., Johnson, V.J., Burleson, G.R., Burleson, F.G.,
EFSA [European Food Safety Authority], 2018. EFSA CONTAM Panel (EFSA Panel on Fletcher, T., 2014. Influenza vaccine response in adults exposed to
Contaminants in the Food Chain), Knutsen HK, Alexander J, Barregard L, Bignami M, perfluorooctanoate and perfluorooctanesulfonate. Toxicol. Sci. 138, 76–88. https://
Bruschweiler B, Ceccatelli S, Cottrill B, Dinovi M, Edler L, Grasl-Kraupp B, Hogstrand doi.org/10.1093/toxsci/kft269.
C, Hoogenboom LR, Nebbia CS, Oswald IP, Petersen A. R. FSA J. 16, 5194. https:// Martí, A., Marcos, A., Martínez, J.A., 2001. Obesity and immune function relationships.
doi.org/10.2903/j.efsa.2018.5194. Obes. Rev. 2, 131–140. https://doi.org/10.1046/j.1467-789x.2001.00025.x.
Emmett, E.A., Zhang, H., Shofer, F.S., Freeman, D., Rodway, N.V., Desai, C., Shaw, L.M., Matilla-Santander, N., Valvi, D., Lopez-Espinosa, M.J., Manzano-Salgado, C.B.,
2006. Community exposure to perfluorooctanoate: Relationships between serum Ballester, F., Ibarluzea, J., Santa-Marina, L., Schettgen, T., Guxens, M., Sunyer, J.,
levels and certain health parameters. J. Occup. Environ. Med. 48, 771–779. https:// Vrijheid, M., 2017. Exposure to perfluoroalkyl substances and metabolic outcomes in
doi.org/10.1097/01.jom.0000233380.13087.37. pregnant women: Evidence from the Spanish INMA birth cohorts. Environ. Health
Fitz-Simon, N., Fletcher, T., Luster, M.I., Steenland, K., Calafat, A.M., Kato, K., Perspect. 125, 117004 https://doi.org/10.1289/EHP1062.
Armstrong, B., 2013. Reductions in serum lipids with a 4-year decline in serum Olsen, G.W., Burris, J.M., Burlew, M.M., Mandel, J.H., 2003. Epidemiologic assessment
perfluorooctanoic acid and perfluorooctanesulfonic acid. Epidemiology 24, of worker serum perfluorooctanesulfonate (PFOS) and perfluorooctanoate (PFOA)
569–576. https://doi.org/10.1097/EDE.0b013e31829443ee. concentrations and medical surveillance examinations. J. Occup. Environ. Med. 45,
Flaherty, J.M., Connolly, P.D., Decker, E.R., Kennedy, S.M., Ellefson, M.E., Reagen, W.K., 260–270. https://doi.org/10.1097/01.jom.0000052958.59271.10.
Szostek, B., 2005. Quantitative determination of perfluorooctanoic acid in serum and Oulhote, Y., Shamim, Z., Kielsen, K., Weihe, P., Grandjean, P., Ryder, L.P., Heilmann, C.,
plasma by liquid chromatography tandem mass spectrometry. J. Chromatogr. B 2017. Children’s white blood cell counts in relation to developmental exposures to
Anal. Technol. Biomed. Life Sci. 819, 329–338. https://doi.org/10.1016/j. methylmercury and persistent organic pollutants. Reprod. Toxicol. 68, 207–214.
jchromb.2005.03.002. https://doi.org/10.1016/j.reprotox.2016.08.001.
Frisbee, S.J., Brooks, A.P., Maher, A., Flensborg, P., Arnold, S., Fletcher, T., Steenland, K., Park, L.P., Margolick, J.B., Giorgi, J.V., Ferbas, J., Bauer, K., Kaslow, R., Muñoz, A.,
Shankar, A., Knox, S.S., Pollard, C., Halverson, J.A., Vieira, V.M., Jin, C., Leyden, K. 1992. Influence of HIV-1 infection and cigarette smoking on leukocyte profiles in
M., Ducatman, A.M., 2009. The C8 Health Project: Design, methods, and homosexual men. The Multicenter AIDS Cohort Study. J. Acquir. Immune Defic.
participants. Environ. Health Perspect. 117, 1873–1882. https://doi.org/10.1289/ Syndr. 5, 1124–1130.
ehp.0800379. Peden-Adams, M.M., Keller, J.M., Eudaly, J.G., Berger, J., Gilkeson, G.S., Keil, D.E.,
Gaylord, A., Berger, K.I., Naidu, M., Attina, T.M., Gilbert, J., Koshy, T.T., Han, X., 2008. Suppression of humoral immunity in mice following exposure to
Marmor, M., Shao, Y., Giusti, R., Goldring, R.M., Kannan, K., Trasande, L., 2019. perfluorooctane sulfonate. Toxicol. Sci. 104, 144–154. https://doi.org/10.1093/
Serum perfluoroalkyl substances and lung function in adolescents exposed to the toxsci/kfn059.
World Trade Center disaster. Environ. Res. 172, 266–272. https://doi.org/10.1016/ Pedersen, K.M., Çolak, Y., Ellervik, C., Hasselbalch, H.C., Bojesen, S.E., Nordestgaard, B.
j.envres.2019.02.024. G., 2019. Smoking and increased white and red blood cells. Arterioscler. Thromb.
George-Gay, B., Parker, K., 2003. Understanding the complete blood count with Vasc. Biol. 39, 965–977. https://doi.org/10.1161/ATVBAHA.118.312338.
differential. J. Perianesthesia Nurs. 18, 96–117. https://doi.org/10.1053/ Qin, X. Di, Qian, Z. (Min), Dharmage, S.C., Perret, J., Geiger, S.D., Rigdon, S.E., Howard,
jpan.2003.50013. S., Zeng, X.W., Hu, L.W., Yang, B.Y., Zhou, Y., Li, M., Xu, S.L., Bao, W.W., Zhang, Y.
Giorgi, J.V., Cheng, H.L., Margolick, J.B., Bauer, K.D., Ferbas, J., Waxdal, M., Schmid, I., Z., Yuan, P., Wang, J., Zhang, C., Tian, Y.P., Nian, M., Xiao, X., Chen, W., Lee, Y.L.,
Hultin, L.E., Jackson, A.L., Park, L., Taylor, J.M.G., 1990. Quality control in the flow Dong, G.H., 2017. Association of perfluoroalkyl substances exposure with impaired
cytometric measurement of T-lymphocyte subsets: The Multicenter AIDS Cohort lung function in children. Environ. Res. 155, 15–21. https://doi.org/10.1016/j.
Study experience. Clin. Immunol. Immunopathol. 55, 173–186. https://doi.org/ envres.2017.01.025.
10.1016/0090-1229(90)90096-9. R Core Team, 2020. R: A language and Environment for Statistical Computing. R
Grandjean, P., Andersen, E.W., Budtz-Jørgensen, E., Nielsen, F., Mølbak, K.R., Weihe, P., Foundation for Statistical Computing. Austria, Vienna.
Heilmann, C., 2012. Serum vaccine antibody concentrations in children exposed to Schenker, E.L., Hultin, L.E., Bauer, K.D., Ferbas, J., Margolick, J.B., Giorgi, J.V., 1993.
perfluorinated compounds. JAMA 307, 391–397. https://doi.org/10.1001/ Evaluation of a dual-color flow cytometry immunophenotyping panel in a
jama.2011.2034. multicenter quality assurance program. Cytometry 14, 307–317. https://doi.org/
10.1002/cyto.990140311.

9
M.-J. Lopez-Espinosa et al. Environment International 156 (2021) 106599

Son, H.Y., Lee, S., Tak, E.N., Cho, H.S., Shin, H.I., Kim, S.H., Yang, J.H., 2009. [WWW Document]. accessed 6.8.20. https://ntp.niehs.nih.gov/ntp/ohat/pfoa_pf
Perfluorooctanoic acid alters T lymphocyte phenotypes and cytokine expression in os/pfoa_pfosmonograph_508.pdf.
mice. Environ. Toxicol. 24, 580–588. https://doi.org/10.1002/tox.20459. Van Leeuwen, S.P.J., Karrman, A., Van Bavel, B., De Boer, J., Lindstrom, G., 2006.
Sopori, M., 2002. Effects of cigarette smoke on the immune system. Nat. Rev. Immunol. Struggle for quality in determination of perfluorinated contaminants in
2, 372–377. https://doi.org/10.1038/nri803. environmental and human samples. Environ. Sci. Technol. 40, 7854–7860. https://
Steenland, K., Jin, C., MacNeil, J., Lally, C., Ducatman, A., Vieira, V., Fletcher, T., 2009. doi.org/10.1021/es061052c.
Predictors of PFOA levels in a community surrounding a chemical plant. Environ. Watkins, D.J., Josson, J., Elston, B., Bartell, S.M., Shin, H.M., Vieira, V.M., Savitz, D.A.,
Health Perspect. 117, 1083–1088. https://doi.org/10.1289/ehp.0800294. Fletcher, T., Wellenius, G.A., 2013. Exposure to perfluoroalkyl acids and markers of
Sunderland, E.M., Hu, X.C., Dassuncao, C., Tokranov, A.K., Wagner, C.C., Allen, J.G., kidney function among children and adolescents living near a chemical plant.
2019. A review of the pathways of human exposure to poly- and perfluoroalkyl Environ. Health Perspect. 121, 625–630. https://doi.org/10.1289/ehp.1205838.
substances (PFASs) and present understanding of health effects. J. Expo. Sci. Yang, Q., Xie, Y., Eriksson, A.M., Nelson, B.D., DePierre, J.W., 2001. Further evidence for
Environ. Epidemiol. 29, 131–147. https://doi.org/10.1038/s41370-018-0094-1. the involvement of inhibition of cell proliferation and development in thymic and
Tryphonas, H., 2001. Approaches to detecting immunotoxic effects of environmental splenic atrophy induced by the peroxisome proliferator perfluoroctanoic acid in
contaminants in humans. Environ. Health Perspect. 109, 877–884. https://doi.org/ mice. Biochem. Pharmacol. 62, 1133–1140. https://doi.org/10.1016/S0006-2952
10.1289/ehp.01109s6877. (01)00752-3.
United States Department of Health and Human Services-National Toxicology Program Zhu, Y., Qin, X. Di, Zeng, X.W., Paul, G., Morawska, L., Su, M.W., Tsai, C.H., Wang, S.Q.,
[USDHHS-NTP], 2016. Systematic review of immunotoxicity associated with Lee, Y.L., Dong, G.H., 2016. Associations of serum perfluoroalkyl acid levels with T-
exposure to perfluorooctanoic acid (PFOA) or perfluorooctane sulfonate (PFOS) helper cell-specific cytokines in children: By gender and asthma status. Sci. Total
Environ. 559, 166–173. https://doi.org/10.1016/j.scitotenv.2016.03.187.

10

You might also like