Next Article in Journal
Effects of a Clinically Indicated Peripheral Intravenous Replacement on Indwelling Time and Complications of Peripheral Intravenous Catheters in Pediatric Patients: A Randomized Controlled Trial
Next Article in Special Issue
Damage to the Testicular Structure of Rats by Acute Oral Exposure of Cadmium
Previous Article in Journal
Regulation/Non-Regulation/Dys-Regulation of Health Behavior, Psychological Reactance, and Health of University Undergraduate Students
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Perfluoroalkyl Chemicals and Male Reproductive Health: Do PFOA and PFOS Increase Risk for Male Infertility?

1
Department of Environmental and Public Health Sciences, University of Cincinnati Medical Center, Cincinnati, OH 45267, USA
2
Center of Environmental Genetics, University of Cincinnati Medical Center, Cincinnati, OH 45267, USA
3
Cincinnati Cancer Center, University of Cincinnati, Cincinnati, OH 45267, USA
*
Author to whom correspondence should be addressed.
Int. J. Environ. Res. Public Health 2021, 18(7), 3794; https://doi.org/10.3390/ijerph18073794
Submission received: 30 December 2020 / Revised: 18 March 2021 / Accepted: 23 March 2021 / Published: 5 April 2021

Abstract

:
Poly- and perfluoroalkyl substances (PFAS) are manmade synthetic chemicals which have been in existence for over 70 years. Though they are currently being phased out, their persistence in the environment is widespread. There is increasing evidence linking PFAS exposure to health effects, an issue of concern since PFAS such as perfluorooctane sulfonate (PFOS) and perfluorooctanoic acid (PFOA) bioaccumulate in humans, with a half-life of years. Many epidemiological studies suggest that, worldwide, semen quality has decreased over the past several decades. One of the most worrying effects of PFOS and PFOA is their associations with lower testosterone levels, similar to clinical observations in infertile men. This review thus focuses on PFOS/PFOA-associated effects on male reproductive health. The sources of PFAS in drinking water are listed. The current epidemiological studies linking increased exposure to PFAS with lowered testosterone and semen quality, and evidence from rodent studies supporting their function as endocrine disruptors on the reproductive system, exhibiting non-monotonic dose responses, are noted. Finally, their mechanisms of action and possible toxic effects on the Leydig, Sertoli, and germ cells are discussed. Future research efforts must consider utilizing better human model systems for exposure, using more accurate PFAS exposure susceptibility windows, and improvements in statistical modeling of data to account for the endocrine disruptor properties of PFAS.

1. Introduction

Poly- and perfluoroalkyl substances (PFAS) comprise more than 3000 individual compounds and generally contain a carbon chain backbone of 4 to 14 atoms in length and a charged functional moiety, such as a sulfonate or carboxylate [1]. Of all the long-chain PFAS family members, perfluorooctane sulfonate (PFOS) and perfluorooctanoic acid (PFOA) are the most studied PFAS. PFAS have been manufactured for over 70 years [1], and though they are currently being phased out in the US, their persistence in the environment is widespread and concerning to human health. Additional reasons for concern are reports of bioaccumulation of PFAS in humans and wildlife and increasing evidence linking environmentally relevant exposures to health effects. Both PFOS and PFOA have a substantial database of epidemiological, pharmacokinetic, toxicological, and mechanistic studies [2,3,4,5,6,7,8]. The chemical and thermal stability of a perfluoroalkyl moiety, and its hydrophobic and lipophobic nature, lead to its incorporation into surfactants, additives, and polymers by a variety of industries [9,10]. The carbon–fluorine (C-F) bond, being the strongest covalent bond, gives PFAS its thermal stability [11]. PFOS and PFOA have an eight-carbon-atom backbone. They have been used in an array of formulations, in firefighting foams, in hydraulic fluids for aircraft, and for waterproofing textiles and non-stick household cookware [1,12,13]. The present PFAS contamination within the environment is the result of consumer–product degradation, firefighting activities, and discharges from PFAS manufacturing facilities [14].
Epidemiological studies have shown that human PFOA and PFOS exposure is probably linked with increased risk for thyroid disease, high cholesterol, ulcerative colitis, kidney, testicular and prostate cancer, and pregnancy-induced hypertension [15,16,17]. In addition, PFAS has been found associated with adverse effects on the immune [18], endocrine, metabolic, and reproductive systems (including fertility and pregnancy outcomes) [19,20,21,22]. However, the weight of evidence supporting these associations varies by outcome, dose of PFAS, and the specific PFAS examined. Although many biological systems are adversely impacted by PFAS, this review focuses only on PFOS/PFOA-associated effects on male reproductive health, and how they may increase susceptibility to male infertility. Epidemiological studies suggest that semen quality has decreased over the past several decades in different countries throughout the world (reviewed in Gabrielsen and Tanrikut, 2016 [23]). The incidence of testicular cancer has also increased in the past 50 years [24,25]. Some studies have shown a strong association between male subfertility and subsequent risk of testicular cancer [26,27,28], suggesting common etiologic factors for infertility and testicular cancer.
One of the most worrying effects of PFOS/PFOA is their effect on Leydig cells in the rat testis, causing Leydig cell hyperplasia [29,30] and lower testosterone levels [31,32], similar to clinical observations in infertile men [33,34,35,36,37,38,39,40]. In our review, rodent studies are examined for common threads and possible endocrine disruptor activities of PFAS. Reports from animal and epidemiological studies implicate PFAS as an endocrine disruptor [15,34,36,41,42]. Endocrine disruptors are exogenous chemicals that can function as agonists or antagonists of hormones, thus disrupting hormone signaling or production [43]. Organs which depend on a constant influx of hormones for proper functioning, such as the testis and ovary, are especially vulnerable to the endocrine disruptor effects of PFAS, predominantly during windows of susceptibility, such as fetal development, infancy, puberty, and aging. Because of this, several epidemiological and rodent studies examine associations between neonatal and pubertal PFAS exposure and male reproductive health outcomes, as detailed later in this review. Early-life exposure to endocrine disruptors has been associated with epigenetic reprogramming, which manifests effects later in life, to increase the risk of disease [44,45]. In this context, we discuss the role of neonatal or pubertal exposures to environmental PFAS in increasing the incidence of reduced sperm counts [37,46] in adults.
In summary, this review discusses the various sources of exposure to PFOS/PFOA, leading to their high concentrations in the general population and in occupational groups. Levels of PFAS exposure in the developing fetus, and with the corresponding birth weight and size, is examined as a separate section, as gestational exposures have wide implications for increasing the risk of male infertility at adulthood. Evidence for sex differences resulting in differential PFOS/PFOA exposure in rodents and in humans is examined as a variable affecting the suitability of rodents as models for PFOS/PFOA exposures. The epidemiological and rodent studies are examined keeping in mind the non-monotonic (non-linear) dose response [47,48,49] usually exhibited by endocrine disruptors. Challenges, data-gaps, and areas for further research are addressed towards the end of each section.

2. Exposure Sources and Concentrations in the Population

PFAS are found in wastewater, as well as in rivers and lakes. They are present in drinking water, storm water, and groundwater [12,50,51,52,53,54,55]. The PFAS not removed in wastewater treatment plants and, consequently, released into the environment are a concern because of the possible presence of these compounds in water used for drinking and recreation. This existence of PFAS in drinking water is an alarming trend, since PFAS persist in humans, with a half-life of several years. Today, PFAS are ubiquitous in the serum of almost all US residents and in populations worldwide [56,57,58]. Continued exposure to even relatively low concentrations in drinking water can substantially increase total human exposure due to its bioaccumulation in serum.
Treated wastewater used for irrigation, or released into rivers, has been linked to PFAS in groundwater [59] and in rivers [60]. This appears to occur because conventional wastewater treatment plants are ineffective at PFAS degradation because of their highly stable covalent C-F bond, their low concentration in water, and their high hydrophilicity [12,61]. Biological treatment can break the C-C bond backbone, but this leads to the formation of short-chain PFAS. This may result in the concentration of PFAS being higher in treated water than in untreated water.
Wastewater facilities which incorporate a dual-filter design, carefully monitor for breakthrough, and frequently change filters have found that granular activated carbon filtration is highly effective at removing PFOA [57]. Using the West Virginia/Ohio Valley cohort, researchers have reported decreases in water PFOA from 1.9 to 4.9 ng/mL (parts per billion) to non-quantifiable (<0.016 ng/mL) or non-detectable levels [57] within 1 year. This was accompanied by a decrease in serum PFOA concentration (after water filtration) by ~26% per year, indicating a median serum PFOA half-life of 2.3 years [57]. This study contrasts with a study by Olsen et al. (2007) [62], who estimated that the geometric mean serum elimination half-life of PFOA was 3.8 (95% CI, 3.1–4.4) years for a group of 26 retired workers with previous occupational exposure. Results utilizing a German cohort of 354 men, women, and children showed declines from baseline serum PFOA concentrations of 10–20% per year after filtration of drinking water [54]. However, these apparent contradictions probably reflect differences in sources of drinking water, whether bottled, well-water, or from rivers [57,63], and possibly also additional sources of PFAS exposure inherent in lifestyle choices [55,64,65,66] which the population may favor. In fact, one of the more intriguing observations emerging from the West Virginia/Ohio Valley cohort study was the significant effect of homegrown vegetable consumption on the apparent half-life of PFOA [57].
The primary exposure source for the general population, apart from drinking water, is diet [55,64,65] and the inhalation of PFAS-contaminated indoor air and dust (from furnishings) [66]. PFAS has been detected in 100% of people tested [67,68,69,70], with the exposure of Americans to PFAS ranging from an average of 0.9 ng/mL to over 100 ng/mL. However, populations in Ronneby, Sweden, where up to one third of households were exposed to PFAS-contaminated drinking water, reportedly have PFOS and PFOA concentrations in their blood serum as high as 1500 ng/mL and 92 ng/mL, respectively [71]. In the US, West Virginia/Ohio Valley residents tested from 2005 to 2006 who drank PFAS-contaminated water close to a chemical plant had a mean level of serum PFOA and PFOS of 80 ng/mL (range 0.25–17,556.6 ng/mL) and 22 ng/mL (range 0.25–759.2 ng/mL), respectively [72]. This serum level did not distinguish between residents and factory workers.
Examples of PFAS-exposed occupational groups include the military and firefighters using flame-retardant foams [73,74], World Trade Center first responders [75], and PFAS manufacturing workers [63,76]. The PFAS factory workers appear to have the highest PFAS levels. Occupational exposure in factory workers has shown blood PFOA levels ranging from 2.25 μg/mL in DuPont workers in China [77] to around 100 μg/mL in 3M factory workers [36]. Olsen et al., 2007 [62] examined the serum of retired PFAS production workers and found mean serum levels of PFOS at 800 ng/mL (range, 145–3490 ng/mL) and of PFOA at 691 ng/mL (range, 72–5100 ng/mL). A recent study examining occupational exposure to PFAS and serum levels of PFOS and PFOA in an aging population (55–74 years, n = 154) from upstate New York [78] showed 25% higher PFOS and 80% higher PFOA levels in study participants compared to the National Health and Nutrition Examination Survey (NHANES). Among these, workers with high cumulative workplace exposure had 34% higher PFOS levels compared to the group without occupational exposure [78].
In 2016, the United States Environmental Protection Agency (US EPA) established drinking water Health Advisories of 70 ng/L (70 parts per trillion) for individual and total concentrations of PFOA and PFOS [79]. However, these are non-regulatory advisories and are presently deemed to be insufficiently protective. Current research focuses on the use of strong anion-exchange resins, which seem to remove both long- and short-chain PFAS. However, since their adsorption capacity for short-chain PFAS is lower than that for long-chain PFAS, the removal of short-chain PFAS is more difficult. Further, in the “real” world, there is usually organic matter mixed with the PFAS. Hence, studies on PFAS removal should incorporate experimental conditions which more closely mimic the environmental milieu. The rapid breakdown of PFAS into short-chain PFAS and the repeated regeneration of PFAS-exhausted adsorbents are other factors which require consideration and more research. Newer techniques such as advanced reduction processes also show promise in the removal of PFAS from water [80]. More details concerning research on the removal of PFAS from water by adsorption are reported in the literature [11].

3. Fetal Exposure to PFOS/PFOA Correlate with Changes in Birth Weight and Size

The very desirable chemical properties of PFAS are, ironically, responsible for their environmental persistence and long half-lives in humans. PFOA and PFOS accumulate in the serum, liver, lungs, brain, and kidney, in humans [81]; they are not easily metabolized, with half-life values of PFOS and PFOA in humans ranging from 2.3 to 5.4 years [54,57,62,82]. Studies in rats have shown PFAS accumulation in the testes [83,84] as well as these other organs.
Mice with gestational PFOA exposure show increased placenta weight, reduced fetal –placental weight ratios, and the presence of placental lesions, suggesting a role for placental toxicity [4,85]. Since PFAS can pass through the placenta to the fetus [86,87,88,89], exposure to PFAS starts from the womb. Infants are a particularly vulnerable subpopulation for PFOA’s developmental effects [4]. Their exposure from the mother drinking contaminated water during pregnancy, through breast milk after birth, and/or from formula prepared with contaminated drinking water has been reported [4,63,76,85,86,87,88,89,90]. In a cross-sectional study of singleton deliveries in the US, cord serum samples (n = 293) were analyzed for PFOS and PFOA. PFOA was detected in 100% and PFOS was detected in greater than 99% of cord blood serum samples [56]. Both PFOS and PFOA concentrations were found to be negatively associated with birth weight and size. However, a study utilizing the Danish National Birth Cohort with 1400 mother–infant pairs showed that only maternal plasma PFOA levels correlated with reduced birth weight [58]. A third study [91] incorporating data collected from 428 mother–infant pairs in Sapporo, Japan, indicated that in utero exposure to low levels of PFOS (measured from maternal serum) negatively correlated with birth weight in female infants only, with PFOA showing no such association. The inconsistencies in these three reports could be due to the timeframe of serum collection or the overall range of concentrations of PFOS/PFOA in the blood. For example, Fei et al. (2007) [58] reported that PFOS and PFOA levels in maternal plasma were, on average, 35.3 ng/mL (range, 6.4 to 106.7 ng/mL) and 5.6 ng/mL (range, <1.0 to 41.5 ng/mL), respectively. Apelberg et al. 2007 [56] reported that the median cord serum PFOS and PFOA levels were 5 ng/mL (range, <0.2 to 34.8 ng/mL) and 1.6 ng/mL (range, 0.3 to 7.1 ng/mL), respectively, while Washino, 2009 [91] showed average PFOS and PFOA levels in maternal serum to be 5.6 ng/mL (range, 1.3 to 16.2 ng/mL) and 1.4 ng/mL (range, <0.5 to 5.3 ng/mL), respectively.
These reports of embryonic exposure to PFOS and PFOA causing changes in birth body weight are a concern because these chemicals may not only directly disrupt fetal development of the male reproductive organs [32,92,93] but may have effects on steroidogenic hormone levels and sperm parameters at adulthood [31,32,46]. As outlined in the Barker Hypothesis [44,45] on the developmental origin of disease, an endocrine disruptor is capable of inducing epigenetic changes (discussed in modes of action) in different organ systems; some of these changes may remain hidden until adulthood, when they increase the risk for disease.

4. Studies Linking Environmentally Relevant Exposure to Male Reproductive Health

4.1. Sex Differences in Elimination Rates in Rats and Higher PFOS/PFOA Levels in Males in the Human General Population

In the rat, not only is the elimination half-life of PFOA shorter than that in monkeys and humans, but a notable sex difference exists [94,95,96,97,98], with females excreting the PFOA much more rapidly (half-life (t1/2) less than 1 day) than males (t1/2 = 15 days). One study found that testosterone affected the PFOA’s urinary elimination and was thus a key determinant of the sex difference in PFOA elimination in rats [99]. The transporter molecules (organic anion transporter) OAT2 and OAT3 were found to be responsible for PFOA transport in rat kidneys [99]. OAT2 mRNA levels in male rats were only 13% of that in female rats and were negatively regulated by testosterone and positively regulated by estrogen. This sex difference in the elimination of PFOA contrasts with the serum half-life of PFOA in mice, monkeys, and humans, which, while different, did not show sex-dependent differences in elimination (reviewed in Post et al., 2012 [100]). However, as noted by Olsen et al., 2008 [101] using the American Red Cross database, the geometric mean plasma concentration for PFOS was statistically significantly higher for males (17.1 ng/mL) than females (12.3 ng/mL). The geometric mean plasma concentration for PFOA in the general human population was 3.4 ng/mL (95% CI 3.3−3.6) but significantly higher for males (3.9 ng/mL) than females (3.0 ng/mL). Support for these sex-based differences have also been observed in the NHANES database [67]. This argues for a mechanism by which human males retain higher levels of PFOA and PFOS in their system. There is only one human study from Ronneby, Sweden, where a marked sex difference was noted with more rapid PFOS elimination in women but only marginally for PFOA [71]. The fact that the half-life of PFOA and PFOS in humans is much longer (measured in years) than in rodents (measured in hours or days), and the renal clearance in humans is negligible, contrary to the active excretion in animals, makes human risk assessment based on animal experiments problematic for these chemicals. However, the higher levels of PFAS in human and rat males than in females, as well as the ability to control for exogenous factors when performing animal studies, makes rodent studies essential to understand the mechanism underlying PFAS action in testes.

4.2. Epidemiological Evidence Linking PFOS and PFOA to Human Male Reproductive Health

The relation of PFAS to various fertility parameters, such as levels of reproductive hormones and sperm quality and levels, has been examined in many epidemiologic studies.
Epidemiological evidence shows that PFOA and PFAS exert significant effects on male reproduction parameters. Workers at the 3M company (Saint Paul, MN, USA), where PFOA was produced, had a 10% increase in mean estradiol levels observed among employees who had the highest levels of serum PFOA. However, this association was confounded by body mass index and became non-significant [102]. A cross-sectional study on 212 exposed males from the Veneto region, Italy, found that increased levels of PFOA (but not PFOS) in serum and seminal fluids positively correlated with circulating testosterone and luteinizing hormone (LH), with a resultant reduction in semen quality, testicular volume, penile length, and anogenital distance [34]. A cross-sectional study from Nanjing, China [103], on 664 adult men showed that seminal PFOA and PFOS levels were significantly associated with a lower percentage of progressive sperm and higher percentage of DNA fragmentation. Use of serum PFOA and PFOS levels made their association weaker compared to the levels in seminal fluids [103]. A follow-up study [33] using the same population confirmed the association between serum and semen PFOA levels with semen parameters and showed a decrease in total testosterone and free testosterone. A study in the US [69] examining semen quality among 501 male partners of couples planning pregnancy showed a correlation between PFOA and PFOS exposure and semen quality, with PFOA and PFOS being associated with a lower percentage of sperm with coiled tails. A study using 588 partners of pregnant women from Greenland, Poland, and Ukraine found that, across countries, sperm concentration, total sperm count, and semen volume were not consistently associated with PFOS or PFOA levels. The most consistent findings were inverse associations between PFOS exposure and sperm morphology [104]. A positive association between PFOA and semen motility [104] and between PFOA and sperm DNA damage [105] was not consistently found across countries. While one cannot rule out that this is a chance finding due to multiple statistical tests being performed, it is also quite likely that different mixtures of PFAS are present in differing environments, which influences the final associations with male reproductive parameters. In support of our supposition, a study utilizing 105 Danish men [37] reported that higher exposure to mixtures of PFOA and PFOS was associated with a decrease in sperm count and number of morphologically normal sperm, as opposed to a weaker association of these sperm parameters with PFOA alone. Moreover, though the highest PFOS–PFOA exposure group displayed a lower total testosterone level, it was not statistically significant. A follow-up cross-sectional study of a cohort of 247 men 5 years later by the same group [38] showed that PFOS levels were negatively associated with testosterone, but earlier findings of decreased sperm morphology were not replicated. As discussed in their publication, this discrepancy could possibly be due to a lack of highly exposed individuals, meaning that the limits of the first study would make all the individuals in the second cohort fall into the “low PFAS” category.
Vested et al., 2013 [106] examined the effects of in utero exposure to PFOA and PFOS in 169 male offspring (19–21 years of age) from a pregnancy cohort established in Aarhus, Denmark, in 1988–1989. They found trends of lower sperm concentration, sperm count, as well as higher FSH (follicle-stimulating hormone) and LH, with higher in utero exposure to PFOA. Lopez-Espinosa et al., 2016 [107] studied 1169 boys (6–9 years of age) living near a chemical plant in the Mid-Ohio Valley (US) with local contamination from PFOA in drinking water. For this cohort, a significant inverse association was found between PFOA levels and testosterone levels, and between PFOS levels with estradiol, testosterone, and IGF-1 (Insulin-like Growth Factor 1) levels. Similarly, a study with 540 Taiwanese subjects aged 12–30 years showed that serum concentrations of PFOA and PFOS were negatively associated with serum levels of sex hormone-binding globulin (SHBG), FSH, and testosterone [108].
In contrast to the above studies showing various degrees of association between PFOS/PFOA and male reproductive parameters, some other groups have found that PFAS exposure is not significantly associated with either semen quality or reproductive hormone levels [39,102,109,110]. The reason for these apparent contradictions could be: (i) composition and concentration levels of various PFAS within the exposure mixture; (ii) age of cohort; (iii) racial differences; (iv) differences in diet; and/or (v) susceptibility windows of exposure. A detailed summary table of the effects of PFOS and PFOA exposure on serum parameters and hormone levels can be found in a review by Bach et al., 2016 [3].
In conclusion, epidemiological studies point to human PFOS/PFOA exposure being associated with deleterious male reproductive outcomes and changes in hormone levels of estrogen, testosterone, LH, FSH, and SHBG. The endocrine-disrupting property of PFOS/PFOA makes associations between PFOS/PFOA levels and outcomes especially problematic, since, instead of a linear response shown by most toxic compounds, endocrine disruptors usually exhibit non-monotonic dose responses. This might, in part, be why some studies did not show dose outcomes expected of that population. It is especially interesting that the three studies [106,107,108] examining either gestational or pubertal PFAS exposure showed more consistent PFAS–testosterone associations, pointing to the exposure during developmental windows of susceptibility as being a key factor influencing reproductive outcomes. To determine the mechanism underlying the cause–effects, an examination of the results from rodent and in vitro studies is required.

4.3. Rodent Studies Linking PFOS and PFOA to Male Reproductive Health

The reproductive effects of PFOA have been studied in rats, mice, monkeys, and rabbits. The rat does not appear to be a suitable animal model since it shows a sex-dependent half-life of PFOA. Moreover, testosterone appears to inhibit the renal clearance of PFOA [97,99] in rats. Male and female mice exhibit an estimated half-life of 12–20 days and may be a better model system, especially for PFOA (reviewed in Post et al., 2012 [100]). Indeed, we found that, except for one study using 11–12-week-old CD rats, PFOA had no effects on reproductive hormone levels or on male reproductive health (Table 1; [29]).
PFOS and PFOA have resulted in dose-dependent developmental effects in rats and mice involving birth weight, postnatal survival, and postnatal growth in surviving animals [32,36,92,93,108,111,112,113,114,115,116]. The most common change with possible adverse effects on male reproduction on exposure to PFOS and PFOA appears to be fluctuations in hormone levels (Table 1, Table 2, Table 3 and Table 4). As described in Table 1, Table 2, Table 3 and Table 4 in more detail, for some studies, the levels of either serum or testicular testosterone appeared to decrease [32,36,92,93,108,111,112,113,114,115,116]; in others, the level of estrogen increased [36,117]. However, this was not a universal phenomenon as some studies show opposite effects, no changes in these two hormone levels [115,118,119,120], or dose-dependent changes [112]. Similarly, levels of FSH and LH displayed non-consistent effects. The receptors for hormones [121,122,123] including androgen, estrogen, and FSH were found to change following exposure. As described in Table 3 and Table 4, changes in steroidogenic enzyme levels have been reported [93,116] for PFOS exposure, which could be due to loss of Leydig cell function [30,113].
Examination of impaired spermatogenesis and reduced sperm counts has also shown variable results (refer to Table 1, Table 2, Table 3 and Table 4), with no obvious common thread linking the differences observed. The inconsistencies could be due to several factors:
(i)
Lack of experimental verification of levels of PFOS and PFOA in the serum make it hard to interpret results from rodent studies. Most studies have estimated values of PFOS/PFOA based on previous studies (reviewed by Olsen et al., 2009 [129]). In most cases, PFOS/PFOA accumulation levels in the testes are ignored.
(ii)
Strain differences within the tested rodent population are an added complication. Studies with endocrine disruptors have shown that some strains of rats and mice are more susceptible to endocrine disruptors than others (discussed in Ashby, 2001 [130]).
(iii)
There is very little information on the food/diet fed to these rodents. It has been shown that the fatty acid content of diets could be an important source of experimental variation impacting spermatogenesis [23,131,132,133,134,135,136,137,138].

4.4. Challenges and Areas of Future Research

There is growing evidence to show that PFAS may act as endocrine disruptors on the reproductive system. The endocrine disruptor function of a chemical depends on its concentration within the system and can cause fluctuations in responses and non-monotonic responses, an inherent problem for both human epidemiological studies and for animal studies. Additionally, exposure levels during “windows of susceptibility”, when the organ is more susceptible to hormonal effects, may give more consistent dose outcome results, rather than at the disease endpoint. For example, the epidemiological studies [106,107,108] examining either gestational or pubertal PFAS exposure showed more consistent PFAS dose–testosterone-level associations. Similarly, PFAS exposure pre-puberty and during puberty could be an important determinant to show more consistent associations between PFAS, testosterone levels, and sperm counts or impaired spermatogenesis in rodents. However, there are many other challenges to studying PFAS-related human health effects. These include the lack of an unexposed control population, exposure to PFAS in the form of complex mixtures of variable constituents with little knowledge regarding the potential synergistic effects of mixtures of PFAS, the window of exposure susceptibility impacting PFAS-related adverse health outcomes not being known (gestational, neonatal, puberty, old age, chronic long-term), and interspecies (for rodents) and ethnic differences (humans) regarding toxicological and toxicokinetic effects of PFAS not being fully understood and further complicated by possible sex differences (reviewed by Blake and Fenton, 2020 [4]). Moreover, the reproductive health effects of long-term chronic exposure to low concentrations of PFAS on the endocrine system, including the thyroid, require further examination.

5. Mechanism of Action of PFOS and PFOA

PFOS and PFOA function as endocrine disruptors on the reproductive system. Animal and in vitro studies have shown that levels of testosterone decrease on increasing exposure to PFAS [32,92]. A decrease in gonadal and/or serum testosterone or increase in estrogen skews the testosterone/estrogen ratio, leading to hormonal imbalance. Aromatase is the key enzyme responsible for converting testosterone to estrogen. In support, the use of aromatase inhibitors in infertile men with low testosterone/estrogen ratios improved both hormonal and semen parameters (reviewed in Schlegel et al., 2012 [40]). Within the testis, the Leydig cells and Sertoli cells work together to sustain and regulate the development of the germ cells into the mature spermatozoa.

5.1. Leydig Cells

Leydig cells produce and secrete testicular testosterone, which accounts for over 95% of circulatory testosterone [35,139]. In addition to aromatase, PFAS exposure has been revealed to be significantly associated with the expression of key enzymes involved in the steroidogenesis process, thereby implying that inhibition of steroidogenic enzyme activity within Leydig cells, or a delay in the maturation/death of Leydig cells, may be a contributing factor to the effects that PFAS exert on androgen secretion in the testis. Steroidogenic acute regulatory protein (STARD1) regulates cholesterol transfer within the mitochondria, which is followed by the conversion of cholesterol to pregnenolone by CYP11A1 (cytochrome P450 family 11A1). Moreover, 17β-Hydroxysteroid dehydrogenase (HSD17B) is involved in the inter-conversion of dehydroepiandrosterone (DHEA) and androstenediol, androstenedione and testosterone, and estrone and estradiol [140]. Rodent and in vitro studies with human or rodent Sertoli cells, Leydig cells, spermatogonia, or seminiferous tubule sections have found that PFOA or PFOS decrease the levels of steroidogenic enzymes such as progesterone, STARD1, CYP11A1, HSD3B1, HSD17B, HSD11B1, and CYP17A1 [92,111,116,123,141]. Additionally, the expression levels of testicular receptors for gonadotropin, growth hormone, and IGF-1 were considerably reduced in mice exposed daily to PFOS [116].

5.2. Sertoli Cells

Sertoli cells secrete androgen-binding protein in the lumen of tubules under the action of FSH [142]. They form tight junctions with each other and maintain the blood–testis barrier responsible for protecting the differentiating germ cell from the immune system. It has been shown that PFOS induces rapid disorganization of the actin- and microtubule-based cytoskeleton in primary cultures of rodent and human Sertoli cells, a phenomenon mediated by RAC-alpha serine/threonine-protein kinase (Akt1/2) [143]. As a result, the blood–testis barrier is destabilized, resulting in male reproductive dysfunction. We found that the exposure of mouse Sertoli cells to PFOA results in a dose-dependent decrease in cell survival (Figure 1a). This observation agrees with data from PFOS- and PFOA-exposed mice, where Sertoli cells showed evidence of morphologic injuries such as the presence of vacuoles, absence of Sertoli cells (“ghost” cells), and disorganization of the cell layers within the seminiferous tubules (probably due to loss of Sertoli cells). [111,127].

5.3. Germ Cells

Spermatogenesis is the process by which germ cells within the seminiferous tubules undergo mitotic and meiotic divisions to form the haploid spermatids, which differentiate and mature into spermatozoa and finally the mature sperm. The germ cells express androgen receptors (AR), estrogen receptor beta, as well as aromatase, and thus are constantly responding to the hormonal milieu surrounding them. Any changes in the hormone levels or receptor expression could result in impaired spermatogenesis. In male pups gestationally exposed to Bisphenol A, our laboratory experiments have shown loss of estrogen receptor beta protein in round spermatids and increased aromatase expression on the acrosomes of the developing spermatids [131,132]. A more thorough investigation of the expression of nuclear receptors estrogen receptor beta, AR, and aromatase in the PFOS/PFOA-exposed dividing, differentiating, and maturing germ cells, as well as Leydig and Sertoli cells within the seminiferous tubule, is thus lacking.
Our research (Figure 1) has found that the effects of PFOA exposure on germ cells are more complex and depend on the stage of the differentiating germ cells. In mouse spermatogonia cell lines, exposure to PFOA (Figure 1b) resulted in an increase in cell proliferation and survival at lower concentrations of PFOA and a decrease in/inhibition of cell proliferation at higher concentrations. For spermatocyte cell lines (cells at pre-meiotic stage), PFOA exposure resulted in a dose-dependent decrease in cell survival (Figure 1c). These results are worrying and require further investigation, since any perturbations in the growth and differentiation of germ cells is thought to result in dysregulation of spermatogenesis.

5.4. Prospective Areas of Investigation into Modes of Action

Identification of early epigenetic biomarkers for PFOS/PFOA exposure which correlate to adverse male reproductive health outcomes could result in improved monitoring of disease susceptibility in a population. Further examination is thus required into epigenetic mechanisms such as histone modifications, DNA methylation, changes in non-coding microRNA, and RNA modifications as modes of action for PFOS/PFOA in the testis. Lu et al., 2007 [144] identified differentially expressed miRNAs in mouse testes after PFOA exposure and showed that miR-133b-3p targets clathrin light chain A, which is involved in endocytosis and thus could be responsible for testicular toxicity. While PFOA and PFOS have been shown to cause global DNA methylation and differentially alter histone modifications in breast cancer cells [145,146,147], this mechanism has not been explored for cells in the testis. Recently, m(6)A demethylase ALKBH5 was shown to regulate spermatogenesis [148]. PFOA and PFOS modification of the expression of this enzyme, and of RNA epigenetics, is a fairly new field of investigation [148] which needs additional inquiry.

6. Looking to the Future

A more fast-track method of testing the potency of these chemicals for human male reproductive health is necessary. Organ culture/ex vivo culture, allografting human testicular fragments under the skin of sexually mature, castrated, immune-deficient mice, and germ cell transportation to rete testis of mice are some of the currently used investigative methods. A fourth method being developed is the establishment of an in vitro human testis model using human testicular cells including human Sertoli, Leydig, and germ cells, similar to mouse or rat 3-D-based testis models [149]. Going one step further, integrating these models with a high throughput method for the monitoring of environmental toxicants’ effects on male reproductive dysfunction (including sperm morphology, sperm counts, and sperm DNA integrity) would be desirable. These methods will also serve as a platform for a future mechanistic understanding of spermatogenesis.
Although the use of PFOS and PFOA has been reduced due to their health impact, the total amount of PFAS introduced into the environment has not reduced because the long-chain compounds have been replaced by short and ultra-short PFAS [53]. While short-chain PFAS potentially bioaccumulate less compared to long-chain, they are still environmentally persistent [11,150]. Their biological effects are still relatively unknown, and they are more recalcitrant to clean-up attempts. Many agencies, such as The Center for Disease Control (CDC), Agency for Toxic Substances and Disease Registry (ATSDR), the US Environmental Protection Agency (EPA), the World Health Organization (WHO), International Agency for Research on Cancer (IARC), and the National Toxicology Program (NTP), have recorded the adverse effects of PFOS and PFOA [18,19,20,77,98,151]. However, these reports only address the possible health effects associated with legacy PFAS, and there are little to no data on health outcomes associated with replacement PFAS. This needs to be rectified.

7. Summary

PFOS/PFOA contamination within the environment is of concern because of their long half-life in humans and their environmental persistence. Epidemiological and rodent studies suggest associations between increased PFAS exposure and changes in the levels of some key steroidogenic enzymes [92,111,116,123,141,152,153] including gonadal or serum testosterone, testicular receptors for gonadotropin, growth hormone, and IGF-1 [116] and semen parameters. The variation and inconsistencies in some effects of PFAS exposure could be explained on the basis of PFOS/PFOA being endocrine disruptors exhibiting non-monotonic responses, or animal model systems being insensitive to hormonal fluctuations. This is a recurrent problem inherent to endocrine disruptors [49] and new statistical approaches to address this issue need to applied. Moreover, examination of the epigenetic programming (miRNA, DNA methylation, and RNA and histone modifications) triggered by PFOS/PFOA during development should be considered as a highly sensitive end-point to monitor for biomarkers of exposure, because epigenetics is a key mechanism for developmental plasticity.

Funding

This study was supported in part by grants from the National Institutes of Health (P30ES006096) and the Department of Defense (W81XWH-15-1-0353 (PT)).

Institutional Review Board Statement

Not Applicable.

Informed Consent Statement

Not Applicable.

Data Availability Statement

No Additional Data to report.

Acknowledgments

We thank Jimmy Antia for his technical assistance.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Buck, R.C.; Franklin, J.; Berger, U.; Conder, J.M.; Cousins, I.T.; de Voogt, P.; Jensen, A.A.; Kannan, K.; Mabury, S.A.; van Leeuwen, S.P. Perfluoroalkyl and polyfluoroalkyl substances in the environment: Terminology, classification, and origins. Integr. Environ. Assess. Manag. 2011, 7, 513–541. [Google Scholar] [CrossRef]
  2. Bach, C.C.; Bech, B.H.; Brix, N.; Nohr, E.A.; Bonde, J.P.; Henriksen, T.B. Perfluoroalkyl and polyfluoroalkyl substances and human fetal growth: A systematic review. Crit. Rev. Toxicol. 2015, 45, 53–67. [Google Scholar] [CrossRef]
  3. Bach, C.C.; Vested, A.; Jørgensen, K.T.; Bonde, J.P.; Henriksen, T.B.; Toft, G. Perfluoroalkyl and polyfluoroalkyl substances and measures of human fertility: A systematic review. Crit. Rev. Toxicol. 2016, 46, 735–755. [Google Scholar] [CrossRef]
  4. Blake, B.E.; Fenton, S.E. Early life exposure to per- and polyfluoroalkyl substances (PFAS) and latent health outcomes: A review including the placenta as a target tissue and possible driver of peri- and postnatal effects. Toxicology 2020, 443, 152565. [Google Scholar] [CrossRef]
  5. Foresta, C.; Tescari, S.; Di Nisio, A. Impact of perfluorochemicals on human health and reproduction: A male’s perspective. J. Endocrinol. Investig. 2018, 41, 639–645. [Google Scholar] [CrossRef]
  6. Gebbink, W.A.; van Leeuwen, S.P.J. Environmental contamination and human exposure to PFASs near a fluorochemical production plant: Review of historic and current PFOA and GenX contamination in the Netherlands. Environ. Int. 2020, 137, 105583. [Google Scholar] [CrossRef] [PubMed]
  7. Klaunig, J.E.; Hocevar, B.A.; Kamendulis, L.M. Mode of Action analysis of perfluorooctanoic acid (PFOA) tumorigenicity and Human Relevance. Reprod. Toxicol. 2012, 33, 410–418. [Google Scholar] [CrossRef] [PubMed]
  8. Luz, A.L.; Anderson, J.K.; Goodrum, P.; Durda, J. Perfluorohexanoic acid toxicity, part I: Development of a chronic human health toxicity value for use in risk assessment. Regul. Toxicol. Pharm. 2019, 103, 41–55. [Google Scholar] [CrossRef]
  9. Kissa, E. Fluorinated Surfactants: Synthesis–Properties–Applications; Marcel Dekker: New York, NY, USA, 1994; p. 469. [Google Scholar]
  10. Kissa, E. Fluorinated Surfactants and Repellents, 2nd ed.; Revised and Expanded; Marcel Dekker: New York, NY, USA, 2001; p. 640. [Google Scholar]
  11. Gagliano, E.; Sgroi, M.; Falciglia, P.P.; Vagliasindi, F.G.A.; Roccaro, P. Removal of poly- and perfluoroalkyl substances (PFAS) from water by adsorption: Role of PFAS chain length, effect of organic matter and challenges in adsorbent regeneration. Water Res. 2020, 171, 115381. [Google Scholar] [CrossRef]
  12. Rahman, M.F.; Peldszus, S.; Anderson, W.B. Behaviour and fate of perfluoroalkyl and polyfluoroalkyl substances (PFASs) in drinking water treatment: A review. Water Res. 2014, 50, 318–340. [Google Scholar] [CrossRef] [PubMed]
  13. Xiao, F. Emerging poly- and perfluoroalkyl substances in the aquatic environment: A review of current literature. Water Res. 2017, 124, 482–495. [Google Scholar] [CrossRef] [PubMed]
  14. Hu, X.C.; Andrews, D.Q.; Lindstrom, A.B.; Bruton, T.A.; Schaider, L.A.; Grandjean, P.; Lohmann, R.; Carignan, C.C.; Blum, A.; Balan, S.A.; 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. [Google Scholar] [CrossRef]
  15. Crawford, N.M.; Fenton, S.E.; Strynar, M.; Hines, E.P.; Pritchard, D.A.; Steiner, A.Z. Effects of perfluorinated chemicals on thyroid function, markers of ovarian reserve, and natural fertility. Reprod. Toxicol. 2017, 69, 53–59. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Domingo, J.L.; Nadal, M. Per- and Polyfluoroalkyl Substances (PFASs) in Food and Human Dietary Intake: A Review of the Recent Scientific Literature. J. Agric. Food Chem. 2017, 65, 533–543. [Google Scholar] [CrossRef] [PubMed]
  17. Blake, B.E.; Pinney, S.M.; Hines, E.P.; Fenton, S.E.; Ferguson, K.K. Associations between longitudinal serum perfluoroalkyl substance (PFAS) levels and measures of thyroid hormone, kidney function, and body mass index in the Fernald Community Cohort. Environ. Pollut. 2018, 242, 894–904. [Google Scholar] [CrossRef] [PubMed]
  18. National Toxicology Program (NTP). National Toxicology Program Monograph on Immunotoxicity Associated with Exposures to Pfoa and Pfos. 2016. Available online: http://ntp.niehs.nih.gov/ntp/ohat/pfoa_pfos/pfoa_pfosmonograph_508.pdf (accessed on 31 March 2021).
  19. Agency for Toxic Substances and Disease Registry (ATSDR). Per- and Polyfluoroalkyl Substances (PFAS) and Your Health; Agency for Toxic Substances and Disease Registry (ATSDR): Atlanta, GA, USA, 2020. [Google Scholar]
  20. Benbrahim-Tallaa, L.; Lauby-Secretan, B.; Loomis, D.; Guyton, K.Z.; Grosse, Y.; El Ghissassi, F.; Bouvard, V.; Guha, N.; Mattock, H.; Straif, K. Carcinogenicity of perfluorooctanoic acid, tetrafluoroethylene, dichloromethane, 1,2-dichloropropane, and 1,3-propane sultone. Lancet Oncol. 2014, 15, 924–925. [Google Scholar] [CrossRef]
  21. Olsen, G.W.; Ley, C.A. Prostate Cancer and PFOA. J. Occup. Environ. Med. 2015, 57, e60. [Google Scholar] [CrossRef]
  22. Gilliland, F.D.; Mandel, J.S. Mortality among employees of a perfluorooctanoic acid production plant. J. Occup. Med. 1993, 35, 950–954. [Google Scholar] [CrossRef]
  23. Gabrielsen, J.S.; Tanrikut, C. Chronic exposures and male fertility: The impacts of environment, diet, and drug use on spermatogenesis. Andrology 2016, 4, 648–661. [Google Scholar] [CrossRef] [PubMed]
  24. Drevinskaite, M.; Patasius, A.; Kincius, M.; Jievaltas, M.; Smailyte, G. A Population-Based Analysis of Incidence, Mortality, and Survival in Testicular Cancer Patients in Lithuania. Medicina 2019, 55, 552. [Google Scholar] [CrossRef] [Green Version]
  25. Le Cornet, C.; Lortet-Tieulent, J.; Forman, D.; Béranger, R.; Flechon, A.; Fervers, B.; Schüz, J.; Bray, F. Testicular cancer incidence to rise by 25% by 2025 in Europe? Model-based predictions in 40 countries using population-based registry data. Eur. J. Cancer 2014, 50, 831–839. [Google Scholar] [CrossRef]
  26. Hanson, H.A.; Anderson, R.E.; Aston, K.I.; Carrell, D.T.; Smith, K.R.; Hotaling, J.M. Subfertility increases risk of testicular cancer: Evidence from population-based semen samples. Fertil. Steril. 2016, 105, 322–328. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Walsh, T.J.; Croughan, M.S.; Schembri, M.; Chan, J.M.; Turek, P.J. Increased risk of testicular germ cell cancer among infertile men. Arch. Intern. Med. 2009, 169, 351–356. [Google Scholar] [CrossRef] [Green Version]
  28. Møller, H.; Skakkebaek, N.E. Risk of testicular cancer in subfertile men: Case-control study. BMJ 1999, 318, 559–562. [Google Scholar] [CrossRef] [Green Version]
  29. Biegel, L.B.; Liu, R.C.; Hurtt, M.E.; Cook, J.C. Effects of ammonium perfluorooctanoate on Leydig cell function: In vitro, in vivo, and ex vivo studies. Toxicol. Appl. Pharm. 1995, 134, 18–25. [Google Scholar] [CrossRef] [PubMed]
  30. Li, Z.; Li, C.; Wen, Z.; Yan, H.; Zou, C.; Li, Y.; Tian, L.; Lei, Z.; Li, H.; Wang, Y.; et al. Perfluoroheptanoic acid induces Leydig cell hyperplasia but inhibits spermatogenesis in rats after pubertal exposure. Toxicology 2020, 448, 152633. [Google Scholar] [CrossRef]
  31. Wang, Z.; Zhang, T.; Wu, J.; Wei, X.; Xu, A.; Wang, S.; Wang, Z. Male reproductive toxicity of perfluorooctanoate (PFOA): Rodent studies. Chemosphere 2020, 270, 128608. [Google Scholar] [CrossRef]
  32. Zhao, B.; Li, L.; Liu, J.; Li, H.; Zhang, C.; Han, P.; Zhang, Y.; Yuan, X.; Ge, R.S.; Chu, Y. Exposure to perfluorooctane sulfonate in utero reduces testosterone production in rat fetal Leydig cells. PLoS ONE 2014, 9, e78888. [Google Scholar] [CrossRef] [Green Version]
  33. Cui, Q.; Pan, Y.; Wang, J.; Liu, H.; Yao, B.; Dai, J. Exposure to per- and polyfluoroalkyl substances (PFASs) in serum versus semen and their association with male reproductive hormones. Environ. Pollut. 2020, 266, 115330. [Google Scholar] [CrossRef]
  34. Di Nisio, A.; Sabovic, I.; Valente, U.; Tescari, S.; Rocca, M.S.; Guidolin, D.; Dall’Acqua, S.; Acquasaliente, L.; Pozzi, N.; Plebani, M.; et al. Endocrine Disruption of Androgenic Activity by Perfluoroalkyl Substances: Clinical and Experimental Evidence. J. Clin. Endocrinol. Metab. 2019, 104, 1259–1271. [Google Scholar] [CrossRef] [Green Version]
  35. Dong, Q.; Hardy, M.P. Leydig Cell Function in Man. In Male Hypogonadism: Basic, Clinical, and Therapeutic Principles; Winters, S.J., Ed.; Humana Press: Totowa, NJ, USA, 2004; pp. 23–43. [Google Scholar]
  36. Jensen, A.A.; Leffers, H. Emerging endocrine disrupters: Perfluoroalkylated substances. Int. J. Androl. 2008, 31, 161–169. [Google Scholar] [CrossRef]
  37. Joensen, U.N.; Bossi, R.; Leffers, H.; Jensen, A.A.; Skakkebaek, N.E.; Jørgensen, N. Do perfluoroalkyl compounds impair human semen quality? Environ. Health Perspect. 2009, 117, 923–927. [Google Scholar] [CrossRef]
  38. Joensen, U.N.; Veyrand, B.; Antignac, J.P.; Blomberg Jensen, M.; Petersen, J.H.; Marchand, P.; Skakkebæk, N.E.; Andersson, A.M.; Le Bizec, B.; Jørgensen, N. PFOS (perfluorooctanesulfonate) in serum is negatively associated with testosterone levels, but not with semen quality, in healthy men. Hum. Reprod. 2013, 28, 599–608. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Petersen, M.S.; Halling, J.; Jørgensen, N.; Nielsen, F.; Grandjean, P.; Jensen, T.K.; Weihe, P. Reproductive Function in a Population of Young Faroese Men with Elevated Exposure to Polychlorinated Biphenyls (PCBs) and Perfluorinated Alkylate Substances (PFAS). Int. J. Environ. Res. Public Health 2018, 15, 1880. [Google Scholar] [CrossRef] [Green Version]
  40. Schlegel, P.N. Aromatase inhibitors for male infertility. Fertil. Steril. 2012, 98, 1359–1362. [Google Scholar] [CrossRef]
  41. Coperchini, F.; Awwad, O.; Rotondi, M.; Santini, F.; Imbriani, M.; Chiovato, L. Thyroid disruption by perfluorooctane sulfonate (PFOS) and perfluorooctanoate (PFOA). J. Endocrinol. Investig. 2017, 40, 105–121. [Google Scholar] [CrossRef]
  42. White, S.S.; Fenton, S.E.; Hines, E.P. Endocrine disrupting properties of perfluorooctanoic acid. J. Steroid Biochem. Mol. Biol. 2011, 127, 16–26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Schug, T.T.; Janesick, A.; Blumberg, B.; Heindel, J.J. Endocrine disrupting chemicals and disease susceptibility. J. Steroid Biochem. Mol. Biol. 2011, 127, 204–215. [Google Scholar] [CrossRef] [Green Version]
  44. Rosenfeld, C.S. Homage to the ‘H’ in developmental origins of health and disease. J. Dev. Orig. Health Dis. 2017, 8, 8–29. [Google Scholar] [CrossRef] [Green Version]
  45. Barker, D.J. The origins of the developmental origins theory. J. Intern. Med. 2007, 261, 412–417. [Google Scholar] [CrossRef] [PubMed]
  46. Mann, U.; Shiff, B.; Patel, P. Reasons for worldwide decline in male fertility. Curr. Opin. Urol. 2020, 30, 296–301. [Google Scholar] [CrossRef]
  47. Hill, C.E.; Myers, J.P.; Vandenberg, L.N. Nonmonotonic Dose-Response Curves Occur in Dose Ranges That Are Relevant to Regulatory Decision-Making. Dose Response 2018, 16, 1559325818798282. [Google Scholar] [CrossRef]
  48. Lee, D.H. Evidence of the Possible Harm of Endocrine-Disrupting Chemicals in Humans: Ongoing Debates and Key Issues. Endocrinol. Metab. 2018, 33, 44–52. [Google Scholar] [CrossRef] [PubMed]
  49. Zoeller, R.T.; Vandenberg, L.N. Assessing dose-response relationships for endocrine disrupting chemicals (EDCs): A focus on non-monotonicity. Environ. Health 2015, 14, 42. [Google Scholar] [CrossRef] [Green Version]
  50. Appleman, T.D.; Higgins, C.P.; Quinones, O.; Vanderford, B.J.; Kolstad, C.; Zeigler-Holady, J.C.; Dickenson, E.R. Treatment of poly- and perfluoroalkyl substances in U.S. full-scale water treatment systems. Water Res. 2014, 51, 246–255. [Google Scholar] [CrossRef] [PubMed]
  51. Munoz, G.; Labadie, P.; Botta, F.; Lestremau, F.; Lopez, B.; Geneste, E.; Pardon, P.; Devier, M.H.; Budzinski, H. Occurrence survey and spatial distribution of perfluoroalkyl and polyfluoroalkyl surfactants in groundwater, surface water, and sediments from tropical environments. Sci. Total Environ. 2017, 607–608, 243–252. [Google Scholar] [CrossRef] [PubMed]
  52. Arvaniti, O.S.; Stasinakis, A.S. Review on the occurrence, fate and removal of perfluorinated compounds during wastewater treatment. Sci. Total Environ. 2015, 524–525, 81–92. [Google Scholar] [CrossRef] [PubMed]
  53. Ateia, M.; Maroli, A.; Tharayil, N.; Karanfil, T. The overlooked short- and ultrashort-chain poly- and perfluorinated substances: A review. Chemosphere 2019, 220, 866–882. [Google Scholar] [CrossRef] [PubMed]
  54. Holzer, J.; Goen, T.; Rauchfuss, K.; Kraft, M.; Angerer, J.; Kleeschulte, P.; Wilhelm, M. One-year follow-up of perfluorinated compounds in plasma of German residents from Arnsberg formerly exposed to PFOA-contaminated drinking water. Int. J. Hyg. Environ. Health 2009, 212, 499–504. [Google Scholar] [CrossRef]
  55. Fromme, H.; Wöckner, M.; Roscher, E.; Völkel, W. ADONA and perfluoroalkylated substances in plasma samples of German blood donors living in South Germany. Int. J. Hyg. Environ. Health 2017, 220, 455–460. [Google Scholar] [CrossRef]
  56. Apelberg, B.J.; Witter, F.R.; Herbstman, J.B.; Calafat, A.M.; Halden, R.U.; Needham, L.L.; Goldman, L.R. Cord serum concentrations of perfluorooctane sulfonate (PFOS) and perfluorooctanoate (PFOA) in relation to weight and size at birth. Environ. Health Perspect. 2007, 115, 1670–1676. [Google Scholar] [CrossRef] [Green Version]
  57. Bartell, S.M.; Calafat, A.M.; Lyu, C.; Kato, K.; Ryan, P.B.; Steenland, K. Rate of decline in serum PFOA concentrations after granular activated carbon filtration at two public water systems in Ohio and West Virginia. Environ. Health Perspect. 2010, 118, 222–228. [Google Scholar] [CrossRef]
  58. Fei, C.; McLaughlin, J.K.; Tarone, R.E.; Olsen, J. Perfluorinated chemicals and fetal growth: A study within the Danish National Birth Cohort. Environ. Health Perspect. 2007, 115, 1677–1682. [Google Scholar] [CrossRef]
  59. Szabo, D.; Coggan, T.L.; Robson, T.C.; Currell, M.; Clarke, B.O. Investigating recycled water use as a diffuse source of per- and polyfluoroalkyl substances (PFASs) to groundwater in Melbourne, Australia. Sci. Total Environ. 2018, 644, 1409–1417. [Google Scholar] [CrossRef]
  60. Becker, A.M.; Gerstmann, S.; Frank, H. Perfluorooctane surfactants in waste waters, the major source of river pollution. Chemosphere 2008, 72, 115–121. [Google Scholar] [CrossRef]
  61. Ross, I.; McDonough, J.; Miles, J.; Storch, P.; Thelakkat Kochunarayanan, P.; Kalve, E.; Hurst, J.S.; Dasgupta, S.; Burdick, J. A review of emerging technologies for remediation of PFASs. Remediat. J. 2018, 28, 101–126. [Google Scholar] [CrossRef]
  62. Olsen, G.W.; Burris, J.M.; Ehresman, D.J.; Froehlich, J.W.; Seacat, A.M.; Butenhoff, J.L.; Zobel, L.R. Half-life of serum elimination of perfluorooctanesulfonate, perfluorohexanesulfonate, and perfluorooctanoate in retired fluorochemical production workers. Environ. Health Perspect. 2007, 115, 1298–1305. [Google Scholar] [CrossRef] [PubMed]
  63. Hoffman, K.; Webster, T.F.; Bartell, S.M.; Weisskopf, M.G.; Fletcher, T.; Vieira, V.M. Private drinking water wells as a source of exposure to perfluorooctanoic acid (PFOA) in communities surrounding a fluoropolymer production facility. Environ. Health Perspect. 2011, 119, 92–97. [Google Scholar] [CrossRef] [Green Version]
  64. Fromme, H.; Tittlemier, S.A.; Völkel, W.; Wilhelm, M.; Twardella, D. Perfluorinated compounds--exposure assessment for the general population in Western countries. Int. J. Hyg. Environ. Health 2009, 212, 239–270. [Google Scholar] [CrossRef]
  65. Yamada, A.; Bemrah, N.; Veyrand, B.; Pollono, C.; Merlo, M.; Desvignes, V.; Sirot, V.; Marchand, P.; Berrebi, A.; Cariou, R.; et al. Dietary exposure to perfluoroalkyl acids of specific French adult sub-populations: High seafood consumers, high freshwater fish consumers and pregnant women. Sci. Total Environ. 2014, 491–492, 170–175. [Google Scholar] [CrossRef]
  66. Fraser, A.J.; Webster, T.F.; Watkins, D.J.; Strynar, M.J.; Kato, K.; Calafat, A.M.; Vieira, V.M.; McClean, M.D. Polyfluorinated compounds in dust from homes, offices, and vehicles as predictors of concentrations in office workers’ serum. Environ. Int. 2013, 60, 128–136. [Google Scholar] [CrossRef] [Green Version]
  67. Kato, K.; Wong, L.Y.; Jia, L.T.; Kuklenyik, Z.; Calafat, A.M. Trends in exposure to polyfluoroalkyl chemicals in the U.S. Population: 1999-2008. Environ. Sci. Technol. 2011, 45, 8037–8045. [Google Scholar] [CrossRef]
  68. Calafat, A.M.; Kuklenyik, Z.; Reidy, J.A.; Caudill, S.P.; Tully, J.S.; Needham, L.L. Serum concentrations of 11 polyfluoroalkyl compounds in the U.S. population: Data from the national health and nutrition examination survey (NHANES). Environ. Sci. Technol. 2007, 41, 2237–2242. [Google Scholar] [CrossRef]
  69. Louis, G.M.; Chen, Z.; Schisterman, E.F.; Kim, S.; Sweeney, A.M.; Sundaram, R.; Lynch, C.D.; Gore-Langton, R.E.; Barr, D.B. Perfluorochemicals and human semen quality: The LIFE study. Environ. Health Perspect. 2015, 123, 57–63. [Google Scholar] [CrossRef]
  70. Olsen, G.W.; Ellefson, M.E.; Mair, D.C.; Church, T.R.; Goldberg, C.L.; Herron, R.M.; Medhdizadehkashi, Z.; Nobiletti, J.B.; Rios, J.A.; Reagen, W.K.; et al. Analysis of a homologous series of perfluorocarboxylates from American Red Cross adult blood donors, 2000-2001 and 2006. Environ. Sci. Technol. 2011, 45, 8022–8029. [Google Scholar] [CrossRef]
  71. Li, Y.; Fletcher, T.; Mucs, D.; Scott, K.; Lindh, C.H.; Tallving, P.; Jakobsson, K. Half-lives of PFOS, PFHxS and PFOA after end of exposure to contaminated drinking water. Occup. Environ. Med. 2018, 75, 46–51. [Google Scholar] [CrossRef] [Green Version]
  72. Steenland, K.; Jin, C.; MacNeil, J.; Lally, C.; Ducatman, A.; Vieira, V.; Fletcher, T. Predictors of PFOA levels in a community surrounding a chemical plant. Environ. Health Perspect. 2009, 117, 1083–1088. [Google Scholar] [CrossRef]
  73. Dobraca, D.; Israel, L.; McNeel, S.; Voss, R.; Wang, M.; Gajek, R.; Park, J.S.; Harwani, S.; Barley, F.; She, J.; et al. Biomonitoring in California firefighters: Metals and perfluorinated chemicals. J. Occup. Environ. Med. 2015, 57, 88–97. [Google Scholar] [CrossRef] [Green Version]
  74. Rotander, A.; Kärrman, A.; Toms, L.M.; Kay, M.; Mueller, J.F.; Gómez Ramos, M.J. Novel fluorinated surfactants tentatively identified in firefighters using liquid chromatography quadrupole time-of-flight tandem mass spectrometry and a case-control approach. Environ. Sci. Technol. 2015, 49, 2434–2442. [Google Scholar] [CrossRef]
  75. Tao, L.; Kannan, K.; Aldous, K.M.; Mauer, M.P.; Eadon, G.A. Biomonitoring of perfluorochemicals in plasma of New York State personnel responding to the World Trade Center disaster. Environ. Sci. Technol. 2008, 42, 3472–3478. [Google Scholar] [CrossRef]
  76. Emmett, E.A.; Shofer, F.S.; Zhang, H.; Freeman, D.; Desai, C.; Shaw, L.M. Community exposure to perfluorooctanoate: Relationships between serum concentrations and exposure sources. J. Occup. Environ. Med. 2006, 48, 759–770. [Google Scholar] [CrossRef]
  77. US Environmental Protection Agency. Draft Risk Assessment of the Potential Human Health Effects Associated with Exposure to Perfluorooctanoic Acid and Its Salts; US EPA Office of Pollution Prevention Toxics Risk Assessment Division: Washington, DC, USA, 2005; AR2261136.
  78. Tanner, E.M.; Bloom, M.S.; Wu, Q.; Kannan, K.; Yucel, R.M.; Shrestha, S.; Fitzgerald, E.F. Occupational exposure to perfluoroalkyl substances and serum levels of perfluorooctanesulfonic acid (PFOS) and perfluorooctanoic acid (PFOA) in an aging population from upstate New York: A retrospective cohort study. Int. Arch. Occup Environ. Health 2018, 91, 145–154. [Google Scholar] [CrossRef]
  79. Post, G.B. Recent US State and Federal Drinking Water Guidelines for Per- and Polyfluoroalkyl Substances. Environ. Toxicol Chem. 2020, 40, 550–563. [Google Scholar] [CrossRef]
  80. Cui, J.; Gao, P.; Deng, Y. Destruction of Per- and Polyfluoroalkyl Substances (PFAS) with Advanced Reduction Processes (ARPs): A Critical Review. Environ. Sci. Technol. 2020, 54, 3752–3766. [Google Scholar] [CrossRef]
  81. Pérez, F.; Nadal, M.; Navarro-Ortega, A.; Fàbrega, F.; Domingo, J.L.; Barceló, D.; Farré, M. Accumulation of perfluoroalkyl substances in human tissues. Environ. Int. 2013, 59, 354–362. [Google Scholar] [CrossRef]
  82. Stubleski, J.; Salihovic, S.; Lind, P.M.; Lind, L.; Dunder, L.; McCleaf, P.; Eurén, K.; Ahrens, L.; Svartengren, M.; van Bavel, B.; et al. The effect of drinking water contaminated with perfluoroalkyl substances on a 10-year longitudinal trend of plasma levels in an elderly Uppsala cohort. Environ. Res. 2017, 159, 95–102. [Google Scholar] [CrossRef]
  83. Iwabuchi, K.; Senzaki, N.; Mazawa, D.; Sato, I.; Hara, M.; Ueda, F.; Liu, W.; Tsuda, S. Tissue toxicokinetics of perfluoro compounds with single and chronic low doses in male rats. J. Toxicol. Sci. 2017, 42, 301–317. [Google Scholar] [CrossRef] [Green Version]
  84. Cui, L.; Zhou, Q.F.; Liao, C.Y.; Fu, J.J.; Jiang, G.B. Studies on the toxicological effects of PFOA and PFOS on rats using histological observation and chemical analysis. Arch. Environ. Contam. Toxicol. 2009, 56, 338–349. [Google Scholar] [CrossRef] [PubMed]
  85. Blake, B.E.; Cope, H.A.; Hall, S.M.; Keys, R.D.; Mahler, B.W.; McCord, J.; Scott, B.; Stapleton, H.M.; Strynar, M.J.; Elmore, S.A.; et al. Evaluation of Maternal, Embryo, and Placental Effects in CD-1 Mice following Gestational Exposure to Perfluorooctanoic Acid (PFOA) or Hexafluoropropylene Oxide Dimer Acid (HFPO-DA or GenX). Environ. Health Perspect. 2020, 128, 27006. [Google Scholar] [CrossRef] [PubMed]
  86. Verner, M.A.; Ngueta, G.; Jensen, E.T.; Fromme, H.; Völkel, W.; Nygaard, U.C.; Granum, B.; Longnecker, M.P. A Simple Pharmacokinetic Model of Prenatal and Postnatal Exposure to Perfluoroalkyl Substances (PFASs). Environ. Sci. Technol. 2016, 50, 978–986. [Google Scholar] [CrossRef]
  87. Grønnestad, R.; Villanger, G.D.; Polder, A.; Kovacs, K.M.; Lydersen, C.; Jenssen, B.M.; Borgå, K. Maternal transfer of perfluoroalkyl substances in hooded seals. Environ. Toxicol. Chem. 2017, 36, 763–770. [Google Scholar] [CrossRef]
  88. Mamsen, L.S.; Björvang, R.D.; Mucs, D.; Vinnars, M.T.; Papadogiannakis, N.; Lindh, C.H.; Andersen, C.Y.; Damdimopoulou, P. Concentrations of perfluoroalkyl substances (PFASs) in human embryonic and fetal organs from first, second, and third trimester pregnancies. Environ. Int. 2019, 124, 482–492. [Google Scholar] [CrossRef]
  89. Mamsen, L.S.; Jönsson, B.A.G.; Lindh, C.H.; Olesen, R.H.; Larsen, A.; Ernst, E.; Kelsey, T.W.; Andersen, C.Y. Concentration of perfluorinated compounds and cotinine in human foetal organs, placenta, and maternal plasma. Sci. Total Environ. 2017, 596–597, 97–105. [Google Scholar] [CrossRef] [Green Version]
  90. Goeden, H.M.; Greene, C.W.; Jacobus, J.A. A transgenerational toxicokinetic model and its use in derivation of Minnesota PFOA water guidance. J. Expo. Sci. Environ. Epidemiol. 2019, 29, 183–195. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Washino, N.; Saijo, Y.; Sasaki, S.; Kato, S.; Ban, S.; Konishi, K.; Ito, R.; Nakata, A.; Iwasaki, Y.; Saito, K.; et al. Correlations between prenatal exposure to perfluorinated chemicals and reduced fetal growth. Environ. Health Perspect. 2009, 117, 660–667. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Eggert, A.; Cisneros-Montalvo, S.; Anandan, S.; Musilli, S.; Stukenborg, J.B.; Adamsson, A.; Nurmio, M.; Toppari, J. The effects of perfluorooctanoic acid (PFOA) on fetal and adult rat testis. Reprod. Toxicol. 2019, 90, 68–76. [Google Scholar] [CrossRef]
  93. Zhang, H.; Lu, H.; Chen, P.; Chen, X.; Sun, C.; Ge, R.S.; Su, Z.; Ye, L. Effects of gestational Perfluorooctane Sulfonate exposure on the developments of fetal and adult Leydig cells in F1 males. Environ. Pollut. 2020, 262, 114241. [Google Scholar] [CrossRef]
  94. Vanden Heuvel, J.P.; Kuslikis, B.I.; Van Rafelghem, M.J.; Peterson, R.E. Tissue distribution, metabolism, and elimination of perfluorooctanoic acid in male and female rats. J. Biochem. Toxicol. 1991, 6, 83–92. [Google Scholar] [CrossRef]
  95. Hanhijärvi, H.; Ophaug, R.H.; Singer, L. The sex-related difference in perfluorooctanoate excretion in the rat. Proc. Soc. Exp. Biol. Med. 1982, 171, 50–55. [Google Scholar] [CrossRef]
  96. Ylinen, M.; Hanhijärvi, H.; Jaakonaho, J.; Peura, P. Stimulation by oestradiol of the urinary excretion of perfluorooctanoic acid in the male rat. Pharm. Toxicol. 1989, 65, 274–277. [Google Scholar] [CrossRef]
  97. Kudo, N.; Suzuki, E.; Katakura, M.; Ohmori, K.; Noshiro, R.; Kawashima, Y. Comparison of the elimination between perfluorinated fatty acids with different carbon chain length in rats. Chem. Biol. Interact. 2001, 134, 203–216. [Google Scholar] [CrossRef]
  98. Kemper, R.A. Perfluorooctanoic Acid: Toxicokinetics in the Rat. Report ID: DuPont-7473. USEPA Administrative Record AR-226.1499; DuPont Haskell Laboratories: Concord, NE, USA, 2003. [Google Scholar]
  99. Vanden Heuvel, J.P.; Davis, J.W., 2nd; Sommers, R.; Peterson, R.E. Renal excretion of perfluorooctanoic acid in male rats: Inhibitory effect of testosterone. J. Biochem. Toxicol. 1992, 7, 31–36. [Google Scholar] [CrossRef] [PubMed]
  100. Post, G.B.; Cohn, P.D.; Cooper, K.R. Perfluorooctanoic acid (PFOA), an emerging drinking water contaminant: A critical review of recent literature. Environ. Res. 2012, 116, 93–117. [Google Scholar] [CrossRef]
  101. Olsen, G.W.; Mair, D.C.; Church, T.R.; Ellefson, M.E.; Reagen, W.K.; Boyd, T.M.; Herron, R.M.; Medhdizadehkashi, Z.; Nobiletti, J.B.; Rios, J.A.; et al. Decline in perfluorooctanesulfonate and other polyfluoroalkyl chemicals in American Red Cross adult blood donors, 2000–2006. Environ. Sci. Technol. 2008, 42, 4989–4995. [Google Scholar] [CrossRef] [Green Version]
  102. Olsen, G.W.; Gilliland, F.D.; Burlew, M.M.; Burris, J.M.; Mandel, J.S.; Mandel, J.H. An epidemiologic investigation of reproductive hormones in men with occupational exposure to perfluorooctanoic acid. J. Occup. Environ. Med. 1998, 40, 614–622. [Google Scholar] [CrossRef]
  103. Pan, Y.; Cui, Q.; Wang, J.; Sheng, N.; Jing, J.; Yao, B.; Dai, J. Profiles of Emerging and Legacy Per-/Polyfluoroalkyl Substances in Matched Serum and Semen Samples: New Implications for Human Semen Quality. Environ. Health Perspect. 2019, 127, 127005. [Google Scholar] [CrossRef]
  104. Toft, G.; Jönsson, B.A.; Lindh, C.H.; Giwercman, A.; Spano, M.; Heederik, D.; Lenters, V.; Vermeulen, R.; Rylander, L.; Pedersen, H.S.; et al. Exposure to perfluorinated compounds and human semen quality in Arctic and European populations. Hum. Reprod. 2012, 27, 2532–2540. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Specht, I.O.; Hougaard, K.S.; Spanò, M.; Bizzaro, D.; Manicardi, G.C.; Lindh, C.H.; Toft, G.; Jönsson, B.A.G.; Giwercman, A.; Bonde, J.P.E. Sperm DNA integrity in relation to exposure to environmental perfluoroalkyl substances-a study of spouses of pregnant women in three geographical regions. Reprod. Toxicol. 2012, 33, 577–583. [Google Scholar] [CrossRef] [Green Version]
  106. Vested, A.; Ramlau-Hansen, C.H.; Olsen, S.F.; Bonde, J.P.; Kristensen, S.L.; Halldorsson, T.I.; Becher, G.; Haug, L.S.; Ernst, E.H.; Toft, G. Associations of in utero exposure to perfluorinated alkyl acids with human semen quality and reproductive hormones in adult men. Environ. Health Perspect. 2013, 121, 453–458. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Lopez-Espinosa, M.J.; Mondal, D.; Armstrong, B.G.; Eskenazi, B.; Fletcher, T. Perfluoroalkyl Substances, Sex Hormones, and Insulin-like Growth Factor-1 at 6-9 Years of Age: A Cross-Sectional Analysis within the C8 Health Project. Environ. Health Perspect. 2016, 124, 1269–1275. [Google Scholar] [CrossRef]
  108. Tsai, M.S.; Lin, C.Y.; Lin, C.C.; Chen, M.H.; Hsu, S.H.; Chien, K.L.; Sung, F.C.; Chen, P.C.; Su, T.C. Association between perfluoroalkyl substances and reproductive hormones in adolescents and young adults. Int. J. Hyg. Environ. Health 2015, 218, 437–443. [Google Scholar] [CrossRef] [PubMed]
  109. Raymer, J.H.; Michael, L.C.; Studabaker, W.B.; Olsen, G.W.; Sloan, C.S.; Wilcosky, T.; Walmer, D.K. Concentrations of perfluorooctane sulfonate (PFOS) and perfluorooctanoate (PFOA) and their associations with human semen quality measurements. Reprod. Toxicol. 2012, 33, 419–427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Lewis, R.C.; Johns, L.E.; Meeker, J.D. Serum Biomarkers of Exposure to Perfluoroalkyl Substances in Relation to Serum Testosterone and Measures of Thyroid Function among Adults and Adolescents from NHANES 2011-2012. Int. J. Environ. Res. Public Health 2015, 12, 6098–6114. [Google Scholar] [CrossRef]
  111. Zhang, H.; Lu, Y.; Luo, B.; Yan, S.; Guo, X.; Dai, J. Proteomic analysis of mouse testis reveals perfluorooctanoic acid-induced reproductive dysfunction via direct disturbance of testicular steroidogenic machinery. J. Proteome Res. 2014, 13, 3370–3385. [Google Scholar] [CrossRef]
  112. Lai, K.P.; Lee, J.C.; Wan, H.T.; Li, J.W.; Wong, A.Y.; Chan, T.F.; Oger, C.; Galano, J.M.; Durand, T.; Leung, K.S.; et al. Effects of in Utero PFOS Exposure on Transcriptome, Lipidome, and Function of Mouse Testis. Environ. Sci. Technol. 2017, 51, 8782–8794. [Google Scholar] [CrossRef] [PubMed]
  113. Li, L.; Li, X.; Chen, X.; Chen, Y.; Liu, J.; Chen, F.; Ge, F.; Ye, L.; Lian, Q.; Ge, R.S. Perfluorooctane sulfonate impairs rat Leydig cell development during puberty. Chemosphere 2018, 190, 43–53. [Google Scholar] [CrossRef] [PubMed]
  114. López-Doval, S.; Salgado, R.; Pereiro, N.; Moyano, R.; Lafuente, A. Perfluorooctane sulfonate effects on the reproductive axis in adult male rats. Environ. Res. 2014, 134, 158–168. [Google Scholar] [CrossRef] [PubMed]
  115. Song, P.; Li, D.; Wang, X.; Zhong, X. Effects of perfluorooctanoic acid exposure during pregnancy on the reproduction and development of male offspring mice. Andrologia 2018, 50, e13059. [Google Scholar] [CrossRef]
  116. Wan, H.T.; Zhao, Y.G.; Wong, M.H.; Lee, K.F.; Yeung, W.S.; Giesy, J.P.; Wong, C.K. Testicular signaling is the potential target of perfluorooctanesulfonate-mediated subfertility in male mice. Biol. Reprod. 2011, 84, 1016–1023. [Google Scholar] [CrossRef] [Green Version]
  117. Du, G.; Hu, J.; Huang, Z.; Yu, M.; Lu, C.; Wang, X.; Wu, D. Neonatal and juvenile exposure to perfluorooctanoate (PFOA) and perfluorooctane sulfonate (PFOS): Advance puberty onset and kisspeptin system disturbance in female rats. Ecotoxicol. Environ. Saf. 2019, 167, 412–421. [Google Scholar] [CrossRef]
  118. York, R.G.; Kennedy, G.L., Jr.; Olsen, G.W.; Butenhoff, J.L. Male reproductive system parameters in a two-generation reproduction study of ammonium perfluorooctanoate in rats and human relevance. Toxicology 2010, 271, 64–72. [Google Scholar] [CrossRef] [PubMed]
  119. Butenhoff, J.L.; Kennedy, G.L., Jr.; Frame, S.R.; O’Connor, J.C.; York, R.G. The reproductive toxicology of ammonium perfluorooctanoate (APFO) in the rat. Toxicology 2004, 196, 95–116. [Google Scholar] [CrossRef] [PubMed]
  120. Luebker, D.J.; Case, M.T.; York, R.G.; Moore, J.A.; Hansen, K.J.; Butenhoff, J.L. Two-generation reproduction and cross-foster studies of perfluorooctanesulfonate (PFOS) in rats. Toxicology 2005, 215, 126–148. [Google Scholar] [CrossRef] [PubMed]
  121. Qu, J.H.; Lu, C.C.; Xu, C.; Chen, G.; Qiu, L.L.; Jiang, J.K.; Ben, S.; Wang, Y.B.; Gu, A.H.; Wang, X.R. Perfluorooctane sulfonate-induced testicular toxicity and differential testicular expression of estrogen receptor in male mice. Environ. Toxicol. Pharm. 2016, 45, 150–157. [Google Scholar] [CrossRef]
  122. López-Doval, S.; Salgado, R.; Lafuente, A. The expression of several reproductive hormone receptors can be modified by perfluorooctane sulfonate (PFOS) in adult male rats. Chemosphere 2016, 155, 488–497. [Google Scholar] [CrossRef] [PubMed]
  123. Tian, M.; Huang, Q.; Wang, H.; Martin, F.L.; Liu, L.; Zhang, J.; Shen, H. Biphasic effects of perfluorooctanoic acid on steroidogenesis in mouse Leydig tumour cells. Reprod. Toxicol. 2019, 83, 54–62. [Google Scholar] [CrossRef]
  124. Liu, W.; Yang, B.; Wu, L.; Zou, W.; Pan, X.; Zou, T.; Liu, F.; Xia, L.; Wang, X.; Zhang, D. Involvement of NRF2 in Perfluorooctanoic Acid-Induced Testicular Damage in Male Mice. Biol. Reprod. 2015, 93, 41. [Google Scholar] [CrossRef]
  125. Lu, Y.; Luo, B.; Li, J.; Dai, J. Perfluorooctanoic acid disrupts the blood-testis barrier and activates the TNFα/p38 MAPK signaling pathway in vivo and in vitro. Arch. Toxicol. 2016, 90, 971–983. [Google Scholar] [CrossRef]
  126. Luebker, D.J.; York, R.G.; Hansen, K.J.; Moore, J.A.; Butenhoff, J.L. Neonatal mortality from in utero exposure to perfluorooctanesulfonate (PFOS) in Sprague-Dawley rats: Dose-response, and biochemical and pharamacokinetic parameters. Toxicology 2005, 215, 149–169. [Google Scholar] [CrossRef]
  127. Qiu, L.; Zhang, X.; Zhang, X.; Zhang, Y.; Gu, J.; Chen, M.; Zhang, Z.; Wang, X.; Wang, S.L. Sertoli cell is a potential target for perfluorooctane sulfonate-induced reproductive dysfunction in male mice. Toxicol. Sci. 2013, 135, 229–240. [Google Scholar] [CrossRef] [Green Version]
  128. Qiu, L.; Qian, Y.; Liu, Z.; Wang, C.; Qu, J.; Wang, X.; Wang, S. Perfluorooctane sulfonate (PFOS) disrupts blood-testis barrier by down-regulating junction proteins via p38 MAPK/ATF2/MMP9 signaling pathway. Toxicology 2016, 373, 1–12. [Google Scholar] [CrossRef]
  129. Olsen, G.W.; Butenhoff, J.L.; Zobel, L.R. Perfluoroalkyl chemicals and human fetal development: An epidemiologic review with clinical and toxicological perspectives. Reprod. Toxicol. 2009, 27, 212–230. [Google Scholar] [CrossRef]
  130. Ashby, J. Testing for endocrine disruption post-EDSTAC: Extrapolation of low dose rodent effects to humans. Toxicol. Lett. 2001, 120, 233–242. [Google Scholar] [CrossRef]
  131. Tarapore, P.; Hennessy, M.; Song, D.; Ying, J.; Ouyang, B.; Govindarajah, V.; Leung, Y.K.; Ho, S.M. Data on spermatogenesis in rat males gestationally exposed to bisphenol A and high fat diets. Data Brief. 2016, 9, 812–817. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Tarapore, P.; Hennessy, M.; Song, D.; Ying, J.; Ouyang, B.; Govindarajah, V.; Leung, Y.K.; Ho, S.M. High butter-fat diet and bisphenol A additively impair male rat spermatogenesis. Reprod. Toxicol. 2017, 68, 191–199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Rebholz, S.L.; Jones, T.; Herrick, R.L.; Xie, C.; Calafat, A.M.; Pinney, S.M.; Woollett, L.A. Hypercholesterolemia with consumption of PFOA-laced Western diets is dependent on strain and sex of mice. Toxicol. Rep. 2016, 3, 46–54. [Google Scholar] [CrossRef] [Green Version]
  134. Campos-Silva, P.; Furriel, A.; Costa, W.S.; Sampaio, F.J.; Gregorio, B.M. Metabolic and testicular effects of the long-term administration of different high-fat diets in adult rats. Int. Braz. J. Urol. 2015, 41, 569–575. [Google Scholar] [CrossRef] [Green Version]
  135. Schagdarsurengin, U.; Western, P.; Steger, K.; Meinhardt, A. Developmental origins of male subfertility: Role of infection, inflammation, and environmental factors. Semin. Immunopathol. 2016, 38, 765–781. [Google Scholar] [CrossRef]
  136. Campos-Silva, P.; Costa, W.S.; Sampaio, F.J.B.; Gregorio, B.M. Prenatal and/or postnatal high-fat diet alters testicular parameters in adult Wistar Albino rats. Histol. Histopathol. 2018, 33, 407–416. [Google Scholar]
  137. Ghosh, S.; Mukherjee, S. Testicular germ cell apoptosis and sperm defects in mice upon long-term high fat diet feeding. J. Cell Physiol. 2018, 233, 6896–6909. [Google Scholar] [CrossRef] [PubMed]
  138. Nan, Y.; Yi, S.J.; Zhu, H.L.; Xiong, Y.W.; Shi, X.T.; Cao, X.L.; Zhang, C.; Gao, L.; Zhao, L.L.; Zhang, J.; et al. Paternal cadmium exposure increases the susceptibility to diet-induced testicular injury and spermatogenic disorders in mouse offspring. Chemosphere 2020, 246, 125776. [Google Scholar] [CrossRef]
  139. Ye, L.; Su, Z.J.; Ge, R.S. Inhibitors of testosterone biosynthetic and metabolic activation enzymes. Molecules 2011, 16, 9983–10001. [Google Scholar] [CrossRef]
  140. Labrie, F.; Luu-The, V.; Lin, S.X.; Simard, J.; Labrie, C.; El-Alfy, M.; Pelletier, G.; Bélanger, A. Intracrinology: Role of the family of 17 beta-hydroxysteroid dehydrogenases in human physiology and disease. J. Mol. Endocrinol. 2000, 25, 1–16. [Google Scholar] [CrossRef] [Green Version]
  141. Lu, H.; Zhang, H.; Gao, J.; Li, Z.; Bao, S.; Chen, X.; Wang, Y.; Ge, R.; Ye, L. Effects of perfluorooctanoic acid on stem Leydig cell functions in the rat. Environ. Pollut. 2019, 250, 206–215. [Google Scholar] [CrossRef]
  142. Hagenäs, L.; Ritzén, E.M.; Ploöen, L.; Hansson, V.; French, F.S.; Nayfeh, S.N. Sertoli cell origin of testicular androgen-binding protein (ABP). Mol. Cell Endocrinol. 1975, 2, 339–350. [Google Scholar] [CrossRef]
  143. Gao, Y.; Chen, H.; Xiao, X.; Lui, W.Y.; Lee, W.M.; Mruk, D.D.; Cheng, C.Y. Perfluorooctanesulfonate (PFOS)-induced Sertoli cell injury through a disruption of F-actin and microtubule organization is mediated by Akt1/2. Sci. Rep. 2017, 7, 1110. [Google Scholar] [CrossRef] [Green Version]
  144. Lu, Y.; Wang, J.; Guo, X.; Yan, S.; Dai, J. Perfluorooctanoic acid affects endocytosis involving clathrin light chain A and microRNA-133b-3p in mouse testes. Toxicol. Appl. Pharm. 2017, 318, 41–48. [Google Scholar] [CrossRef] [PubMed]
  145. Liu, W.; Irudayaraj, J. Perfluorooctanoic acid (PFOA) exposure inhibits DNA methyltransferase activities and alters constitutive heterochromatin organization. Food Chem. Toxicol. 2020, 141, 111358. [Google Scholar] [CrossRef] [PubMed]
  146. Pierozan, P.; Cattani, D.; Karlsson, O. Perfluorooctane sulfonate (PFOS) and perfluorooctanoic acid (PFOA) induce epigenetic alterations and promote human breast cell carcinogenesis in vitro. Arch. Toxicol. 2020, 94, 3893–3906. [Google Scholar] [CrossRef]
  147. Rashid, F.; Ramakrishnan, A.; Fields, C.; Irudayaraj, J. Acute PFOA exposure promotes epigenomic alterations in mouse kidney tissues. Toxicol. Rep. 2020, 7, 125–132. [Google Scholar] [CrossRef] [PubMed]
  148. Liu, J.; Jia, G. Methylation modifications in eukaryotic messenger RNA. J. Genet. Genom. 2014, 41, 21–33. [Google Scholar] [CrossRef]
  149. Mahmoud, H. Concise review: Spermatogenesis in an artificial three-dimensional system. Stem Cells 2012, 30, 2355–2360. [Google Scholar] [CrossRef] [PubMed]
  150. Brendel, S.; Fetter, É.; Staude, C.; Vierke, L.; Biegel-Engler, A. Short-chain perfluoroalkyl acids: Environmental concerns and a regulatory strategy under REACH. Environ. Sci. Eur. 2018, 30, 9. [Google Scholar] [CrossRef]
  151. Chiu, W.A.; Guyton, K.Z.; Martin, M.T.; Reif, D.M.; Rusyn, I. Use of high-throughput in vitro toxicity screening data in cancer hazard evaluations by IARC Monograph Working Groups. ALTEX 2018, 35, 51–64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Zhang, S.; Mo, J.; Wang, Y.; Ni, C.; Li, X.; Zhu, Q.; Ge, R.S. Endocrine disruptors of inhibiting testicular 3β-hydroxysteroid dehydrogenase. Chem. Biol. Interact. 2019, 303, 90–97. [Google Scholar] [CrossRef]
  153. Zhao, B.; Hu, G.X.; Chu, Y.; Jin, X.; Gong, S.; Akingbemi, B.T.; Zhang, Z.; Zirkin, B.R.; Ge, R.S. Inhibition of human and rat 3beta-hydroxysteroid dehydrogenase and 17beta-hydroxysteroid dehydrogenase 3 activities by perfluoroalkylated substances. Chem. Biol. Interact. 2010, 188, 38–43. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Cell survival after treatment with indicated concentrations of PFOA. Mouse Sertoli TM4 (a), mouse type B spermatogonia GC-1 (b), and mouse pre-meiotic spermatocyte GC-2 (c) cell lines (obtained from ATCC) were plated in triplicate in a 96-well plate at a density of 20,000 cells/well and incubated overnight. Cells were treated with PFOA at 5 different concentrations ranging from 0 µM to 100 µM followed by an incubation period of 3–4 days. Cell proliferation (MTT) assay was performed. Displayed data are from at least two individual experiments, mean ± standard error (SEM). Significance was calculated using one-way ANOVA with Dunnett’s post-test. * p value < 0.05, ** p value < 0.01, *** p value < 0.001.
Figure 1. Cell survival after treatment with indicated concentrations of PFOA. Mouse Sertoli TM4 (a), mouse type B spermatogonia GC-1 (b), and mouse pre-meiotic spermatocyte GC-2 (c) cell lines (obtained from ATCC) were plated in triplicate in a 96-well plate at a density of 20,000 cells/well and incubated overnight. Cells were treated with PFOA at 5 different concentrations ranging from 0 µM to 100 µM followed by an incubation period of 3–4 days. Cell proliferation (MTT) assay was performed. Displayed data are from at least two individual experiments, mean ± standard error (SEM). Significance was calculated using one-way ANOVA with Dunnett’s post-test. * p value < 0.05, ** p value < 0.01, *** p value < 0.001.
Ijerph 18 03794 g001
Table 1. PFOA exposure and male fertility measures in rats.
Table 1. PFOA exposure and male fertility measures in rats.
AuthorSpecies (Age)ExposureDoseOutcome MeasuresSerum Concentration
Iwabushi et al., 2017 [83] Male Wistar rats (6–7 weeks) Mixture of PFAS (perfluorohexanoic acid, PFOA, PFOS, perfluorononanoic acid)1, 5, and 25 μg/L in drinking water daily for 1 month and 3 monthsHormone levels not examined. Testis not examined. Epididymal sperm counts not done.1-month exposure PFOA: ~6.5–160 µg/kg serum volume 3-month exposure PFOA: ~8.9–184 µg/kg serum volume
Biegel et al., 1995 [29]Male CD rats (11–12 weeks)Ammonium PFOA (APFO)25 mg/kg/day APFO for 14 days.Inhibits testosterone release from Leydig cells. Testis not examined.Not determined
Butenhoff et al., 2004 [119] Male and female Sprague Dawley rats (6-weeks) and F1 pups at Post-natal Day (PND) 21 Ammonium PFOA (APFO)1, 3, 10, or 30 mg/kg/day daily by gavage.Fertility and all sperm parameters normal in all generations.Not determined
Cui et al., 2009 [84] Male Sprague Dawley rats (8-weeks) PFOA5 or 20 mg/kg/day daily for 28 days by gavageNo distinct pathological change in testes.Whole blood 39.2 μg/mL (5 mg/kg/day); 58.8 μg/mL (20 mg/kg/day) Testicle 16.7 μg/mL (5 mg/kg/day); 16.8 μg/mL (20 mg/kg/day)
York et al., 2010 [118] Male and female Sprague Dawley rats (6-weeks) and F1 pups at PND21 Ammonium PFOA (APFO)0, 1, 3, 10, or 30 mg/kg/day daily by gavage.Fertility and all sperm parameters normal in all generations.Not determined
Table 2. PFOA exposure and male fertility measures in mice.
Table 2. PFOA exposure and male fertility measures in mice.
AuthorSpecies (Age)ExposureDoseOutcome MeasuresSerum Concentration
Zhang et al., 2014 [111] Male BALB/c mice (6–8 weeks) PFOA0.31, 1.25, 5, and 20 mg/kg/day daily for 28 days by gavage.Reduced testosterone and progesterone in testis. Disruption of spermatogenesis in 5 and 20 mg/kg/d groups. Reduced epididymal sperm count (only 5 mg/kg/d group tested).Testicle 5.37 μg/g (5 mg/kg/d group) and 8.06 μg/g (20 mg/kg/d group)
Liu et al., 2015 [124] Male Kunming mice (8 weeks) PFOA2.5, 5, or 10 mg/kg/day daily for 14 days.Disruption of spermatogenesis. Reduced epididymal sperm count (dose-dependent effects at all doses). Decreased expression of NRF2 (Nuclear Factor, Erythroid 2 Like 2). Inhibition of antioxidant enzymes superoxide dismutase and catalase. Upregulation of p-p53 and BAX (BCL2 Associated X, Apoptosis Regulator) expression and downregulation of BCL-2 (B-Cell CLL/Lymphoma 2) expression in testis.Not determined
Lu et al., 2016 [125]Male BABL/c mice (6–8 weeks)PFOA1.25, 5, or 20 mg/kg/day daily for 28 days by gavage.Disruption of blood–testes barrier and immune privilege observed in all three PFOA groups.Not determined
Song et al., 2018 [115]Female pregnant micePFOA1, 2.5, or 5 mg/kg/day daily by gavageReduced level of testosterone in male offspring on PND 21. Dose-dependent damage to testis. Number of Leydig cells decreased in 2.5 and 5 mg/kg PFOA groups on PND 21 and 70.Not determined
Table 3. PFOS exposure and male fertility measures in rats.
Table 3. PFOS exposure and male fertility measures in rats.
AuthorSpecies (Age)ExposureDoseOutcome MeasuresSerum Concentration
Iwabushi et al., 2017 [83] Male Wistar rats (6–7 weeks) Mixture of PFAS (C6A, PFOA, PFOS, C9A)1, 5, and 25 μg/L in drinking water daily for 1 month and 3 monthsHormone levels not examined. Testis not examined. Epididymal sperm counts not done.1-month exposure PFOS: 1.09–17.2 µg/kg serum volume 3-month exposure PFOS: 2.7–73.7 µg/kg serum volume
Cui et al., 2009 [84] Male Sprague Dawley rats (8 weeks) PFOS5 or 20 mg/kg/day daily for 28 days by gavageNo distinct pathological change in testesWhole blood 72.0 μg/mL (5 mg/kg/day); not determined (20 mg/kg/day), all died within 26 days exposure. Testicle 39.5 μg/mL (5 mg/kg/day); 127 μg/mL (5 mg/kg/day)
López-Doval et al., 2014 [114] Male Sprague Dawley rats (8 weeks) PFOS0.5; 1.0; 3.0; and 6.0 mg/kg/day for 28 days by gavageCirculating levels of LH and testosterone decrease and FSH increase. Disrupts male reproductive axis activity.Not determined
Zhao et al., 2014 [32] Female pregnant Sprague-Dawley rats PFOS5, 20 mg/kg/day from gestational day 11–19 by gavage.Male F1 generation examined at gestational day 20. Decreased testosterone, impaired fetal Leydig cells with reduced number. Decreased expression of genes expressed by Leydig cells including Cyp11A1 and decreased cholesterol levels.Not determined
López-Doval et al., 2016 [122] Male Sprague Dawley rats (8 weeks) PFOS1.0; 3.0; and 6.0 mg/kg/d daily for 28 days by gavagePFOS inhibits both gene and protein expression of FSH receptor and AR at testicular level. Testis not examined.Not determined
Li et al., 2018 [113]Male Sprague Dawley rats (PND35)PFOS5 or 10 mg/kg/day starting PND 35 for 21 days by gavageLowered serum testosterone levels, decreased expression of genes expressed by Leydig cells (Lhcgr (LH/choriogonadotropin receptor), Cyp11a1, and Cyp17a1).Not determined
Li et al., 2018 [113]Male Sprague Dawley rats (4 weeks)PFOS5 or 10 mg/kg/day starting on PND 35 for 21 days by gavage5–10 mg/kg/d reduced epididymal sperm count and serum testosterone levels. Promoted immature Leydig cell apoptosis. 10 mg/kg/d disrupted Leydig cell specific gene expression (LHCGR, CYP11A1, and CYP17A1). Delayed Leydig cell development during puberty.Not determined
Zhang et al., 2020 [93] Female pregnant Sprague-Dawley rats PFOS1 or 5 mg/kg/day from gestational day 5–20 by gavage.Male F1 generation examined at PND1, 35, 90. Decreased serum testosterone levels. Decreased levels of Scarb1 (Scavenger receptor class B type 1), Cyp11a1, Cyp17a1, and Hsd17b3, Dhh (Desert hedgehog homolog), and Sox9 (SRY-related HMG-box). Inhibition of Leydig cell proliferation.Not determined
Luebker et al., 2005 [120,126] Male-female Sprague Dawley rats (6-weeks) and F1 pups PFOS0.1, 0.4, 1.6, and 3.2 mg/kg/day throughout the 2-generational studyFertility and all sperm parameters normal in all generations. Developmental mortality observed for 1.6 and 3.2 mg/kg/day groups.Not determined
Table 4. PFOS exposure and male fertility measures in mice.
Table 4. PFOS exposure and male fertility measures in mice.
AuthorSpecies (Age)ExposureDoseOutcome MeasuresSerum Concentration
Wan et al., 2011 [116]CD1 male mice (8 weeks)PFOS1, 5, or 10 mg/kg/day for 7, 14, or 21 days by gavage.For 10 mg/kg/d, day21: Serum testosterone levels decreased. Epididymal sperm counts decreased. mRNA expression levels of steroidogenic enzymes (i.e., StAR, CYP11A1, CYP17A1, 3beta-HSD, and 17beta-HSD) were reduced.Not determined
Qiu et al., 2013 [127]Male ICR mice (8 weeks)PFOS0.25, 2.5, 25, and 50 mg/kg/day for 28 days by gavage.For ≥ 2.5mg/kg/day: Significant dose-dependent decrease in sperm count. Significant increase in Sertoli cell vacuolization and disruption of spermatogenesis, significant loss in blood–testis barrier.Not determined
Qiu et al., 2016 [128]Male ICR mice (8 weeks)PFOS0.5, 5, and 10 mg/kg/bw daily for 4 weeks by gavageDose-dependent decrease in sperm count. Loss in blood–testis barrier at dose of 5 mg/kg/bw and higherNot determined
Qu et al., 2016 [121]Male C57 mice (6–8 weeks)PFOS0.5 and 10 mg/kg/day for 5 weeks by gavageSerum testosterone levels decreased. Higher incidence of apoptotic cells, and vacuolations observed in spermatogonia, spermatocytes, and Leydig cells for 10 mg/kg/day group.Not determined
Lai et al., 2017 [112]Female pregnant CD-1 mice (6−8 weeks)PFOS0.3 or 3 mg/kg/dayOn PND1, aberration of lipid metabolism, oxidative stress and cell-junction signaling, perturbations of lipid mediators. On PND63 reduction in serum testosterone and epididymal sperm countPFOS in PND1 testis: ~0.6 µg/g (0.3 mg/kg/d group), ~3.8 µg/g (3 mg/kg/d group)
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Tarapore, P.; Ouyang, B. Perfluoroalkyl Chemicals and Male Reproductive Health: Do PFOA and PFOS Increase Risk for Male Infertility? Int. J. Environ. Res. Public Health 2021, 18, 3794. https://doi.org/10.3390/ijerph18073794

AMA Style

Tarapore P, Ouyang B. Perfluoroalkyl Chemicals and Male Reproductive Health: Do PFOA and PFOS Increase Risk for Male Infertility? International Journal of Environmental Research and Public Health. 2021; 18(7):3794. https://doi.org/10.3390/ijerph18073794

Chicago/Turabian Style

Tarapore, Pheruza, and Bin Ouyang. 2021. "Perfluoroalkyl Chemicals and Male Reproductive Health: Do PFOA and PFOS Increase Risk for Male Infertility?" International Journal of Environmental Research and Public Health 18, no. 7: 3794. https://doi.org/10.3390/ijerph18073794

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop