THE JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM ADVANCE ARTICLE: JCEM Andrea Di Nisio, Iva Sabovic, Umberto Valente, Simone Tescari, Maria Santa Rocca, Diego Guidolin, Stefano Dall’Acqua, Laura Acquasaliente, Nicola Pozzi, Mario Plebani, Andrea Garolla, Carlo Foresta The Journal of Clinical Endocrinology & Metabolism Endocrine Society Submitted: August 28, 2018 Accepted: November 01, 2018 First Online: November 06, 2018 Advance Articles are PDF versions of manuscripts that have been peer reviewed and accepted but not yet copyedited. The manuscripts are published online as soon as possible after acceptance and before the copyedited, typeset articles are published. They are posted "as is" (i.e., as submitted by the authors at the modification stage), and do not reflect editorial changes. No corrections/changes to the PDF manuscripts are accepted. Accordingly, there likely will be differences between the Advance Article manuscripts and the final, typeset articles. The manuscripts remain listed on the Advance Article page until the final, typeset articles are posted. At that point, the manuscripts are removed from the Advance Article page. DISCLAIMER: These manuscripts are provided "as is" without warranty of any kind, either express or particular purpose, or non-infringement. Changes will be made to these manuscripts before publication. Review and/or use or reliance on these materials is at the discretion and risk of the reader/user. In no event shall the Endocrine Society be liable for damages of any kind arising references to, products or publications do not imply endorsement of that product or publication. Downloaded from https://academic.oup.com/jcem/advance-article-abstract/doi/10.1210/jc.2018-01855/5158211 by Endocrine Society Main Account user on 26 November 2018 Endocrine disruption of androgenic activity by perfluoroalkyl substances: clinical and experimental evidence The Journal of Clinical Endocrinology & Metabolism; Copyright 2018 DOI: 10.1210/jc.2018-01855 Endocrine disruption of androgenic activity by perfluoroalkyl substances: clinical and experimental evidence Andrea Di Nisio1, Iva Sabovic1, Umberto Valente1, Simone Tescari1, Maria Santa Rocca1 , Diego Guidolin2, Stefano Dall’Acqua3, Laura Acquasaliente4, Nicola Pozzi4, Mario Plebani5, Andrea Garolla1, Carlo Foresta1* 1 2 3 LE Department of Medicine, Operative Unit of Andrology and Medicine of Human Reproduction, University of Padova, Via Giustiniani, 2, 35128 Padova, Italy. Department of Neurosciences, University of Padova, via Gabelli 65, 35128 Padova, Italy. Department of Pharmaceutical Science, University of Padova, via Marzolo 5, 35128 Padova, Italy 5 IC Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine 1100 South Grand Blvd. St. Louis, MO 63104, USA. Department of Medicine, Laboratory Medicine, University of Padova, Via Giustiniani, 2, 35128 Padova, Italy T ORCiD numbers: R 0000-0002-6576-2183 Foresta A Carlo Received 28 August 2018. Accepted 01 November 2018. D V A N C E Background: Considerable attention has been paid to perfluoroalkyl compounds (PFCs) because of their worldwide presence in humans, wildlife, and environment. A wide variety of toxicological effects is well supported in animals, including testicular toxicity and male infertility. For these reasons, the understanding of epidemiological associations and of the molecular mechanisms involved in the endocrine-disrupting properties of PFCs on human reproductive health is a major concern. Objective: To investigate the relationship between PFCs exposure and male reproductive health. Design: This study was performed within a screening protocol to evaluate male reproductive health in high schools. Patients: this is a cross-sectional study on 212 exposed males from the Veneto region, one of the four areas worldwide heavily polluted with PFCs, and 171 non-exposed controls. Main outcome measures: Anthropometrics, seminal parameters and sex hormones were measured in young males from exposed areas, compared with age-matched controls. We also performed biochemical studies in established experimental models. Results: We found that increased levels of PFCs in plasma and seminal fluid positively correlate with circulating T and with a reduction of semen quality, testicular volume, penile length and AGD. Experimental evidence points towards an antagonistic action of PFOA on the binding of T to AR in gene reporter assay, competition assay on AR-coated SPR chip and AR nuclear translocation assay. Discussion: This study documents that PFCs have a substantial impact on human health as they interfere with hormonal pathways, potentially leading to male infertility. A ADVANCE ARTICLE: JCEM THE JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM 4 PFCs exposure leads to an impairment of male reproductive system, which is supported by experimental evidence showing an interference of these chemicals on the binding of testosterone to its receptor. 1 Downloaded from https://academic.oup.com/jcem/advance-article-abstract/doi/10.1210/jc.2018-01855/5158211 by Endocrine Society Main Account user on 26 November 2018 Perfluoroalkyls disruption of androgenic activity The Journal of Clinical Endocrinology & Metabolism; Copyright 2018 DOI: 10.1210/jc.2018-01855 Introduction LE IC T R A E C N A V D Subjects A THE JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM ADVANCE ARTICLE: JCEM Methods This study was performed within the annual screening protocol to evaluate male reproductive health in the high schools of Padova and surroundings (Veneto Region, North-East of Italy). The aim of this screening is to early diagnose possible risk factors and diseases of the male reproductive system. Here, we report the findings of 383 subjects who voluntarily agreed to complete the cross-sectional study between June 2017 and May 2018. Included subjects underwent an accurate medical visit, measure of anthropometric parameters, ultrasound examination of the testes and semen analysis at our medical center. Written informed consent was obtained from all subjects, and the study was approved by the Research Ethics Committee of the University Hospital of Padova (N. 2208P). The investigation was 2 Downloaded from https://academic.oup.com/jcem/advance-article-abstract/doi/10.1210/jc.2018-01855/5158211 by Endocrine Society Main Account user on 26 November 2018 Perfluoroalkyl compounds (PFCs) are a class of organic molecules that are used in many everyday products such as oil and water repellents, coatings for cookware, carpets, and textiles. Their attractive physio-chemical characteristics (i.e., colourless, odourless, high thermal stability, low chemical reactivity and durability), high availability and low cost ensure widespread use in the industry but also drive persistent accumulation into the environment, making them a potential biohazard for human health(1,2). Indeed, PFCs have been found in human fluids and tissues including the brain, placenta, and testis, which are protected by strong selective barriers(3–7). Interestingly, and for unknown reasons, there seems to be a sex-dependent pharmacodynamic profile, with adult males having a much higher tendency to PFCs accumulation and lower clearance(8–11). Exposure pathways and toxicity mechanisms for PFCs are not well characterized, at least in humans (reviewed in (12)). An attractive hypothesis emerges from recent phenomenological studies correlating the dysfunction of the male reproductive system with the environmental levels of PFCs(13). PFCs may be absorbed by the intestine or inhaled and, once in the circulation, they may act as endocrine disruptors (ED) ultimately leading to genital disorders, such as impaired spermatogenesis and reproductive defects, and antiandrogenic-driven conditions, such as testicular dysgenesis syndrome (13), which is an established risk factor for testis cancer(14,15). PFCs could exert their toxicity on the foetus, new-born, as well as during development, especially in teenagers due to alterations in sex hormones biosynthesis. Recent data suggest that in utero exposure to PFCs is associated to lower sperm quality and higher levels of LH and FSH at adulthood(16). Furthermore, by apparently acting as both anti-androgenic and anti-estrogenic molecules, PFCs might also affect the downstream signalling pathways of sex hormones (17,18), down-regulate the hypothalamic-pituitary axis activity and increase testicular toxicity during development(19– 21). The crucial emerging role PFCs as pollutants of water, soil, and air, and their persistent level in males warrant for more investigation on the mechanisms of PFCs toxicity in humans. In this comprehensive study, we tested the hypothesis that human exposure to PFCs drives androgenic dysfunction and deterioration of the male reproductive system by altering the testosterone (T) interaction with its specific androgen receptor (AR). To investigate the relationship between PFCs exposure and clinical alterations, we studied a cohort of 212 exposed young men from the Veneto region, in the North-East of Italy. With Mid-Ohio valley in the USA, Dordrecht area in the Netherlands, and Shandong district in China, the Veneto region is one of the four areas worldwide heavily polluted with PFCs. To fully characterize the antiandrogenic action of PFCs, and the structural and functional interaction between PFCs, AR, and T, we performed biochemical studies in established experimental models. The Journal of Clinical Endocrinology & Metabolism; Copyright 2018 DOI: 10.1210/jc.2018-01855 LE IC N C E A R T Anthropometric and penile measurements included: height, weight, body mass index (BMI), waist circumference, arm span, crown-to-pubis length, penile length and circumference. These parameters are commonly used to suggest severe forms of congenital or pre-pubertal hypogonadism, such as patients with Klinefelter and Kallmann syndrome(24). Every measure was taken three times to the nearest millimeter. Height was accurately taken from the floor to the crown of the head as described in previous studies(25,26). BMI was calculated using the formula weight (kg)/height (m)2. Waist circumference was measured at the midpoint between the superior border of the iliac crest and the lowest rib(27). The arm span was measured as the distance between the tips of the middle fingers with the arms fully extended (28). The pubis-to-floor distance was measured from the upper edge of pubic symphysis to the floor. The crown-to-pubis length was consequently derived as the difference between height and pubis-to-floor distance (29). The penile length was measured as the linear distance along the dorsal side of the penis extending from the lower edge of the pubic bone to the tip of the glans in the flaccid state. The penis circumference was measured at the middle of the shaft (30). All subjects were evaluated by the same two clinicians. The intra-operator variations were in all cases <5%. Testicular volumes were evaluated by ultrasound, using the standard ellipsoid formula (width × height × length × π/6, CV<10%). A Anogenital distance (AGD) D V The AGD was measured as previously described elsewhere(31), from the posterior base of the scrotum to the center of the anus. The participant was placed in a supine, frog-legged position with his thighs at a 45° angle to the examination table. In a subset of 50 randomly chosen patients, AGD measures were repeated twice by the same technician, and then blindly by the second examiner. Interclass correlation coefficients (ICC) were calculated for repeatability estimation within and between examiners. Repeatability was very high within individual (ICC= 0.979, 95% CI= 0.960 - 0.989) and slightly lower across examiners (ICC= 0.932, 95% CI= 0.873 – 0.964). A THE JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM ADVANCE ARTICLE: JCEM Anthropometric measurements Semen collection and analysis Human semen samples were obtained by masturbation after 2–7 days of sexual abstinence and stored in sterile containers. Samples were allowed to liquefy for 30 min at 37°C and were examined for seminal parameters according to WHO criteria(32). Briefly: semen volume was measured by weighing, assuming a semen density of 1.0 g/ml; sperm concentration was evaluated by hemocytometer (Bürker-Türk; Paul Marienfeld GmbH&Co. KG, LaudaKönigshofen, Germany); sperm morphology was identified from semen smears prepared with 10  µl of well-mixed semen, stained with Papanicolaou and assessed using the Tygerberg 3 Downloaded from https://academic.oup.com/jcem/advance-article-abstract/doi/10.1210/jc.2018-01855/5158211 by Endocrine Society Main Account user on 26 November 2018 performed according to the principles of the Declaration of Helsinki. Participants did not receive any reimbursement. Based on geographical distribution of PFCs pollution (22) subjects were then grouped on the basis of their residence. Regional authorities (23) have defined two different zones within the exposed area, based on the degree of pollution: the red area, which is the one with the highest PFCs levels, and the yellow zone, with slightly lower levels, but at risk for the close proximity with the contamination plume and surroundings (22). Among the 383 subjects included in the study, 83 were resident in the yellow zone, 129 in the red zone, and 171 outside the exposed area (green zone). Specific geographical origin is reported in Supplementary figure 1(22). In order to increase the sample size for subsequent statistical analyses, subjects from red and yellow zone were pooled together as a single exposed group, since no difference has emerged between the two areas for the clinical parameters considered (data not shown), except for non-progressive sperm motility and immotile sperm that where mutually different between groups. Subjects from green zone (non-exposed) were considered as control group. The Journal of Clinical Endocrinology & Metabolism; Copyright 2018 DOI: 10.1210/jc.2018-01855 Sex hormones quantification E A R T IC In a subset of patients (50 controls and 50 exposed subjects), PFCs were evaluated in serum and seminal fluid. For serum analyses, cells are removed from plasma by centrifugation for 10 minutes at 2000 x g. Following centrifugation, the liquid component (plasma) was transferred into a clean polypropylene tube. The quantification of PFOA and PFOS was processed on reversed-phase (RP) liquid chromatography coupled with high-resolution mass spectrometry (LC-MS) Agilent Varian 320 (Agilent Technologies, Santa Clara, CA, USA). Briefly, each sample was dissolved in acetonitrile and fixed amounts of the stable isotopelabeled internal standard were added (MPFOA, MPFOS, Wellington Laboratories, Ontario, Canada). To test the analytical response and to optimize calibration curve standard mixture was used at increasing concentrations (PFAC-MXB, Wellington Laboratories) together with isotope-labeled internal standards (MPFOA, MPFOS) at fixed concentrations. This solution was analysed by LC-MS. The different perfluoroalkyl species were identified by comparing the retention time and mass spectra (i.e. m/z value and isotopic pattern). Quantification of each species was calculated using the corresponding calibration curve. AR gene reporter assay D V A N C All the transfections were performed in HeLa cells, obtained from the American Type Culture Collection (ATCC, Manassas, VA, USA) as previously described(33). Briefly, cells were cultivated in Dulbecco's modified Eagle medium (DMEM) (Thermofisher, Waltham, MA, USA), supplemented with 5% fetal bovine serum (Sigma Aldrich, St. Louis, MO, USA), antibiotics, and antimycotics in a humidified incubator at 37°C with 5% CO2. Transient gene expression assay was performed in 96-well plates using Lipofectamine 2000 reagent (Thermofisher, Waltham, MA, USA) and a Dual-Luciferase reporter assay system (Promega Corp, Madison, WI, USA). HeLa cells were grown in 96-well plates and cotransfected at 70% confluence with 100 ng/well of the expression vector for the full-length human AR (pSV-AR0), 100 ng/well of mouse mammary tumor virus (MMTV)-luciferase reporter plasmid and 10 ng/well of pGL4.74 Renilla luciferase (Promega Corp.) (internal control for transfection efficiency). pSV-AR0 and MMTV-Luc plasmids were a kind gift from Prof. Claessens (University of Leuven, Belgium). Twenty-four hours after transfection, media was replaced with fresh DMEM and test chemicals (PFOA and PFOS 1 µM, Wellington Laboratories, Ontario, Canada) in the absence or presence of 10 nM T (positive control, Sigma Aldrich) were added to each well. Flutamide (1 µM, Sigma Aldrich) served as negative control. Treated cells were harvested 24 h later and lysed with lysis buffer of DualLuciferase reporter assay (Promega Corp.). Luciferase activity was measured with a multilabel plate reader (Wallac Victor, Perkin-Elmer, Waltham, MA, USA) and all data were standardized for luciferase activity. Results are shown as the mean ± SD of three independent experiments, each performed in duplicate. A ADVANCE ARTICLE: JCEM THE JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM PFCs quantification in serum and semen by mass-spectrometry LE Blood was collected in the fasting state between 08:00 and 10:00 AM. Serum total T, FSH and LH were evaluated by commercial electrochemiluminescence immunoassay methods (Elecsys 2010; Roche Diagnostics, Mannheim, Germany). For all parameters, the intra- and interassay coefficient of variation were <8% and <10%, respectively. All determinations were performed in duplicate. Surface Plasmon resonance analyses 4 Downloaded from https://academic.oup.com/jcem/advance-article-abstract/doi/10.1210/jc.2018-01855/5158211 by Endocrine Society Main Account user on 26 November 2018 strict criteria. Sperm motility was graded into total (progressive + non-progressive motility) and progressive motility. Total sperm count (volume × sperm concentration) was also calculated. Seminal parameters were available for 211 exposed subjects and 170 controls, since one subject in each group failed to collect semen. The Journal of Clinical Endocrinology & Metabolism; Copyright 2018 DOI: 10.1210/jc.2018-01855 LE IC D V A N C E A R T The clonal strain of mouse MA-10 Leydig cell line used for AR nuclear translocation assay was purchased from ATCC® (CRL-3050™, ATCC, Manassas, Virginia, USA). Cells were used at the second cell passage from original thawing, in order to maintain the phenotype as close as possible to the one claimed by the manufacturer, and handled as previously described(34). Briefly, cells were seeded on 0.1 % gelatine-coated plasticware and maintained in DMEM/F12 medium, pH 7.7 (GIBCO-Invitrogen, Milano, Italy), supplemented with 20 mM Hepes, 15% horse serum, and 50 µg/ml gentamicin. Starved MA10 cells seeded onto glass slides (BD Biosciences, Milano, Italy) and cultured at different concentrations of T (1 to 100 nM, Sigma Aldrich) and PFOA (0.1 to 1 µM, Wellington Laboratories), alone or in combination. After 24 hours, cells were fixed with 4 % paraformaldehyde/PBS solution for 15 min at room temperature and were permeabilized with 1 % Triton X-100/PBS solution for 10 min at room temperature. Furthermore, samples were saturated with 5 % BSA/5 % normal donkey serum in PBS for 30 min and then incubated overnight at 4 °C with Rabbit polyclonal Anti-Androgen receptor antibody (ab74272, Abcam, Cambridge, UK) for further assessment of AR nuclear translocation by the means of relative quantification of fluorescence density. In the negative control, primary antibodies were omitted. The following day, primary immunoreaction was detected by incubation with IgGFITC goat anti-Rabbit secondary antibody (K1715, Santa Cruz Biotechnology, Dallas, TX, USA). Finally, cells were counterstained with DAPI, mounted with antifade buffer, and analyzed with videoconfocal (VICO) fluorescence microscope (Nikon, Firenze, Italy). The nuclear translocation of AR (the intensity of AR signal within the nucleus relative to the total intensity) was quantified in 20-40 cells using the software ImageJ (http://rsbweb.nih.gov/ij/download.html). Statistical analyses All statistics were calculated using SPSS (Version 23; SPSS Inc., Chicago, IL, USA). p values <0.05 were considered as statistically significant. The results were expressed as means ± SD or as medians + interquartile ranges. The Shapiro-Wilk W test for normality was used to check the distributions of the variables; as almost none of parameters was normally distributed (except height, crown-to-pubis, pubis-to-floor and sperm progressive motility), and almost all of log-transformed distributions did not satisfy normality, non-parametric statistics was applied. Mann-Whitney test was used to assess differences between groups in the anthropometric, seminal and hormonal parameters and in in the concentrations of serum e seminal contaminants. Both raw and adjusted p values are reported; adjustment for multiple comparisons was calculated with the Bonferroni-Holm method. Spearman's rank correlation A THE JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM ADVANCE ARTICLE: JCEM AR nuclear translocation assay 5 Downloaded from https://academic.oup.com/jcem/advance-article-abstract/doi/10.1210/jc.2018-01855/5158211 by Endocrine Society Main Account user on 26 November 2018 Surface Plasmon Resonance (SPR) experiments were performed on a BIAcore-S200 instrument (GE-Healthcare, Chicago IL USA) to monitor the interaction between T or PFOA and the AR. The binding domain AF2 of the AR (650-920, Abcam, Cambridge, MA, USA) was covalently immobilized on a CM5 sensor chip using an amine-coupling chemistry. Binding experiments were carried out by injecting increasing concentrations of T (0-1mM; Sigma Aldrich) and PFOA (0-4 M; Wellington Laboratories) at a flow rate of 30µl/min, using 10mM Hepes pH7.4, 0.15M NaCl containing 3% MeOH (V/V) as running buffer. Each cycle consists of 60-sec contact time, followed by 120-sec dissociation and 30-sec pulse with 100mM Hepes pH7.4, as regeneration step. The response units (RU) at the steady state were plotted as a function of [analyte] and the dissociation constant (Kd) was obtained as a fitting parameter of a binding isotherm. Competition experiments were performed in order to investigate the effect of PFOA on T-AR interaction. Solutions of T (250 M) were incubated with different concentrations of PFOA (0-4 µM) for 10 minutes, and then injected over the AR-coated sensor chip. All experiments were performed in triplicate at 25ºC. The Journal of Clinical Endocrinology & Metabolism; Copyright 2018 DOI: 10.1210/jc.2018-01855 LE IC D V A N C E A R T Anthropometrics and seminal measures of the two groups are reported on Table 1. In particular, subjects from the exposed group showed significantly lower mean testicular volume and shorter penile length and AGD, after adjustment for multiple comparisons. Prior to adjustments, also crown-to-pubis and pubis-to-floor distances, and the respective ratio, differed between groups. No significant difference was observed for age and other anthropometric parameters. For what concerns seminal parameters, exposed subjects showed significantly lower sperm progressive motility and normal sperm morphology, together with higher semen pH and immotile sperm (Table 1). In addition to the reduction in semen quality, also lower sperm count was observed in exposed males, in terms of sperm concentration and total count, although not statistically significant after correction for multiple comparisons (Table 1). The overview of seminal and genital alterations is suggestive of an impairment of androgenic signalling in these subjects. To confirm this hypothesis, we evaluated gonadal-pituitary axis in a subset of 100 randomly-chosen subjects (50 from the control group and 50 from the exposed group) that underwent also LC-MS quantification of PFOA and PFOS in serum and seminal plasma, in order to confirm the geographic selection criteria based on exposure patterns. In the subgroup from the exposed area, increased semen pH was confirmed, together with a lower percentage of sperm with normal morphology, reduced penile length and circumference and smaller testicular volume, but not AGD, although only after adjustment for multiple comparisons (Table 2). No significant difference has emerged in terms of other seminal or anthropometric parameters. PFOA was detected in serum from 98% subjects and in 96% of the respective seminal plasma, whereas PFOS was detected in 90% of sera and 86% of seminal plasma. PFCs quantification has confirmed higher serum levels of both PFOA and PFOS in exposed subjects (Table 2), with the former being the prominent species in blood, with a mean of 14.99 ng/mL in the exposed group and 4.71 ng/mL in control subjects. In addition, the concentration of PFOA, but not PFOS, was higher in the in seminal plasma from exposed subjects, although lower than serum levels (Table 2). Hormonal analyses showed higher levels of total T and LH in the exposed group, compared with control counterparts (Table 2). In the correlation analyses, serum and seminal plasma levels of PFOS and PFOA were highly correlated with each other (Spearman’s ρ =0.216, p =0.034 and ρ =0.294, p =0.003, respectively), as were PFOA plasma and semen concentrations (ρ =0.449, p <0.001), but not PFOS plasma and semen levels (ρ 0.163, p =0.111). Serum PFOA levels were positively correlated with total T (ρ =0.305, p =0.002; Figure 1) and LH (ρ =0.224, p =0.046), as were seminal PFOA (ρ =0.346, p <0.001 and ρ =0.259, p =0.021) and with the proportion of sperm A ADVANCE ARTICLE: JCEM THE JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM Results 6 Downloaded from https://academic.oup.com/jcem/advance-article-abstract/doi/10.1210/jc.2018-01855/5158211 by Endocrine Society Main Account user on 26 November 2018 coefficients were calculated to evaluate the correlations between the concentrations of each contaminant and the variables of interest. In the gene reporter assay, to examine differences between treatment groups and positive control (T 10 nM), one-way analysis of variance (ANOVA) was performed with a Dunnett´s post hoc test, given the normal distribution of data. Due to relatively few data points per concentration and non-normality of the data, non-parametric statistics were used to analyse AR nuclear translocation assay. The Kruskal-Wallis test was used to compare differences between concentrations and the Jonckheere-Terpstra test (two-tailed) was used to analyze for a linear trend between concentration and response. If one or both tests showed a significant difference (p <0.05), the Mann–Whitney test with Bonferroni correction was used to compare each concentration with the control. The same procedure was applied to comparisons between different stimuli (PFOA 0.1, 1 and 10 µM and flutamide 1 µM) and T, within each T concentration. The Journal of Clinical Endocrinology & Metabolism; Copyright 2018 DOI: 10.1210/jc.2018-01855 LE IC T R A E C N A V D THE JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM ADVANCE ARTICLE: JCEM A 7 Downloaded from https://academic.oup.com/jcem/advance-article-abstract/doi/10.1210/jc.2018-01855/5158211 by Endocrine Society Main Account user on 26 November 2018 with normal morphology (ρ =-0.303, p =0.002 and ρ =-0.225, p =0.025 respectively). Again, seminal PFOA showed a positive correlation also with pH (ρ =0.203, p=0.042). Both serum and semen PFOA, but not PFOS, were associated with reduced testicular volume (ρ =0.211, p=0.037 and ρ =-0.277, p= 0.006, respectively). Within this framework of clinical signs suggestive of an endocrine disruption of androgen action by PFCs, we aimed to experimentally test the hypothesis of an interference of these chemicals on the AR, the mediator of androgen signalling. To this end, an AR gene reporter assay on HeLa cells transiently co-transfected with an MMTV-LUC reporter vector and an AR expression plasmid pSV-AR0. PFOS and PFOA at a concentration of 1 µM acted as mild agonists of AR (10.5% and 11.6%, Figure 2). Upon co-incubation with T 10 nM, both PFOS and PFOA elicited a significant (p <0.001) antagonistic effect on T-induced activation of AR at concentrations comparable with those reported in highly exposed populations. These compounds antagonized the T-induced response (set to 100 %) down to 73.5 and 64.2 %, respectively (Figure 2), with PFOA being the most potent AR inhibitor. The relative potencies of the tested compounds were approximately twice lower than the inhibitor control flutamide (Figure 2). Given the highest occurrence of PFOA in the serum of exposed Italian populations and its higher potency compared to PFOS in the gene reporter assay, we focused on PFOA to elucidate the anti-androgenic mechanism of PFCs. SPR measurements were performed to monitor the real-time interaction between T and PFOA with the AR. In this experiment, the AF2 binding domain of AR was immobilized on a CM5 sensor chip and solutions of T (0 to 1 mM) and PFOA (0 to 4 µM) were injected separately at different concentrations. Despite the low molecular weight of T (288 Da), SPR resolved the interaction and provided a dissociation constant Kd =174±32 µM (Figure 3A-B). In contrast, no interaction between PFOA (up to 4 µM) and AR was detected under the same experimental conditions (Figure 3C-D). Higher concentrations of PFOA were not tested because of the limited solubility in the running buffer. Next, we performed competition experiment to assess whether the presence of PFOA would reduce the binding of T to AR. We incubated a solution of T with different concentrations of PFOA and the resulting mixture was flowed over the same ARcoated sensor chip. At the highest concentration tested, we observed a small but significant (35%) decrease of T binding suggesting that the presence of PFOA reduces the binding of T to its receptor (Figure 3E). In a final set of experiments, we aimed to test in vitro the putative inhibitory effect of PFOA on AR. To this end, nuclear translocation assay was performed on murine Leydig MA10 cells, cultured at different concentrations of T (1 to 100 nM) and PFOA (0.1 to 1 µM), alone or in combination. Flutamide 1 µM served as negative control. In the positive control, T elicited a significant (all p <0.001 vs unstimulated cells) and consistent AR nuclear internalization, with approximately 90% of positive signal within the nucleus, even at the lowest concentration (Figure 4, 5). By addition of androgen inhibitor flutamide, AR nuclear signal decreased down to 17.9%, 20.9% and 36.4% at T concentrations of 1, 10 and 100 nM respectively. A very low signal was detected in cells incubated with PFOA 0.1, 1 or 10 µM (2.4, 3.4, 7.1%; Figure 4), which however was comparable with that of the negative unstimulated control (4.1%). When Leydig cells were co-incubated with both PFOA and T, a dose-dependent inhibition of AR nuclear translocation was observed for increasing concentrations of PFOA, which was inversely correlated to T concentration (Figure 4, 5): at the highest T concentration (100 nM), PFOA did not affect AR internalization at any concentration, but at physiologically relevant T levels (10 nM), AR nuclear signal significantly decreased at highest PFOA concentration (10 µM). On the other hand, at lower levels of T (1 nM), PFOA induced a significant reduction of AR internalization at any tested concentration (Figure 5). A Jonckheere-Terpstra test for ordered alternatives showed that The Journal of Clinical Endocrinology & Metabolism; Copyright 2018 DOI: 10.1210/jc.2018-01855 Discussion IC T R A E C N A V D A ADVANCE ARTICLE: JCEM THE JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM LE This study documents that PFCs have a substantial impact on human male health as they directly interfere with hormonal pathways potentially leading to male infertility. We found that increased levels of PFCs in plasma and seminal fluid positively correlate with circulating T and with a reduction of semen quality, testicular volume, penile length and AGD. Experimental evidence supports our observational results and points towards an antagonistic action of PFOA on the binding of T to its natural AR. The investigation covered an area of around 150 km2 in the provinces of Vicenza and Padua and to some extent Verona, with 350,000-400,000 people potentially exposed(35,36). These areas are heavily polluted with concentrations of PFCs up to 6872 ng/L for all PFCs and up to 3733 ng/L for PFOA alone in surface waters and up to 3138 ng/l for all PFCs and up to 1886 ng/l for PFOA in drinking water, that is >1000 fold higher than control values (0.5 to 8 ng/L)(37). Compared to median concentrations of PFOA and PFOS in blood serum of the general population in Italy, we found levels of PFOA more than 5 times higher in plasma and semen compared to control. Although slightly inferior to the PFCs levels calculated on 13856 subjects aged 14-40, during a surveillance programme promoted by the Veneto from the most polluted “red zone”(38), our results are consistent with previous findings(35), and our sensitive LC-MS method is able to differentiate between exposed population and controls. Interestingly, the majority of the exposed male population showed a reduction in testicular volume, penile length, and AGD, but not anthropometrics in males aged 18-19. These findings could be explained considering that AGD and anthropometric measures are differentially determined during fetal and pre-pubertal development, respectively(24). Accordingly, genital development is concomitant with AGD determination(39). Therefore we could speculate an hypothetic involvement of PFC to in-utero rather than late ED exposure. Prenatal exposure to androgens during the “masculinization programming window”, a critical window during testicular development, is positively associated with AGD in mammals(39). On these bases, AGD has been suggested as a putative marker of prenatal exposure to chemicals with known anti-androgenic effect, or ED in general. For example, exposure to phthalates(40), dioxins(41) and bisphenol A(42) has been associated with a reduction in AGD. As the first report on water contamination of PFCs goes back to 1977(43), the magnitude of the problem is alarming as it affects an entire generation of young individuals, from 1978 onwards. PFCs toxicity also concerns adult life independently from in-utero exposure. This implies that healthy individuals living in territories contaminated with PFCs could present signs of toxicity. In vitro and animal studies on PFCs toxicity have shown a detrimental effect of PFCs on testicular function due to the alteration of steroidogenic machinery and subsequent defect of spermatogenesis(44–48). Two cross-sectional studies reported negative associations of PFOS, or high PFOA and PFOS combined, with the proportion of morphologically normal spermatozoa in adult men(49,50). This is in agreement with our findings in which we observe a significant reduction in progressive sperm motility in exposed subjects. The exact mechanism, however, is not clear, and possibly involves an impairment of mitochondrial activity, as observed in the endocrine disruptor bisphenol A(51). Another important finding is the association between PFOA and seminal plasma pH, indicative of an interference of PFCs at a prostatic level. The presence of PFCs in seminal plasma reported by previous groups (52) and by us suggests either a prostatic or testicular 8 Downloaded from https://academic.oup.com/jcem/advance-article-abstract/doi/10.1210/jc.2018-01855/5158211 by Endocrine Society Main Account user on 26 November 2018 there was a statistically significant trend of higher AR nuclear translocation scores with increasing concentration of T, alone (z =2.416, p =0.016) or in combination with flutamide (z =3.695, p<0.001) and PFOA, at any tested concentration (all p <0.001). The Journal of Clinical Endocrinology & Metabolism; Copyright 2018 DOI: 10.1210/jc.2018-01855 LE IC T R A E C N A V D A THE JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM ADVANCE ARTICLE: JCEM Acknowledgments: We thank all the staff of the Unity of Andrology and Reproductive Medicine at the University Hospital for the constant support. Data availability All data supporting the findings of this study are available within the article and its Supplementary Information. All other data are available from the corresponding author upon reasonable request. Authors contribution: 9 Downloaded from https://academic.oup.com/jcem/advance-article-abstract/doi/10.1210/jc.2018-01855/5158211 by Endocrine Society Main Account user on 26 November 2018 origin of PFCs, that could explain a weak association of plasma PFOS concentration with incident prostate cancer (53). This aspect, however, requires further investigations. Overall, the inefficient recognition between T and its receptor in the presence of PFCs could explain the clinical symptoms in the exposed individuals. It would also explain why higher levels of T are found in exposed subjects, which is a compensatory mechanism, as supported by increased LH. Interestingly, in the only study that evaluated young males from an exposed pregnancy cohort, prenatal exposure to PFOA was associated later in adult life with lower sperm concentration and total sperm count(16). The same study also reported an alteration of the hypothalamic-pituitary axis, with higher levels of LH and FSH, but not T. Several experiments provide direct evidence that PFOA inhibits the binding of T to AR. First, PFOS and PFOA elicit a significant antagonistic effect (~25%) on testosterone-induced activation of AR in HeLa cells. This result is in agreement with the study by Kjeldsen and colleagues(18), but in contrast with Behr(54) and Du(55), who have used different cell lines, reporter plasmids and co-treatment conditions. Second, PFOA diminishes the binding of T to the purified receptor. Third, PFOA significantly reduces the translocation of AR to the nucleus in murine Leydig cells. Remarkably, co-incubation of physiological concentrations of T in adults (10 nM) and PFOA led to a ∼20% reduction of AR nuclear signal, at concentrations reported in regions with point source drinking water contamination (1 µM)(35,36,56) and in occupationally-exposed fluorochemical workers (10 µM)(57). Despite the convincing biological effect, the mechanism of inhibition remains elusive and requires more biochemical investigations to be unveiled. Moreover, quantification of circulating androgens with more precise methods could unveil further associations with sex steroids, and given the cross-sectional design of the study, further confounding factors could be included, such as socioeconomic status. Because of the partial antagonist effect in our assays, PFOA may act as an allosteric or non-competitive inhibitor thereby blocking dimerization of the receptor and its translocation to the nucleus. This would explain as to why SPR experiments failed to monitor the interaction between PFOA and covalently immobilized monomeric AR. Alternatively, PFOA could interact with T thereby diminishing the concentration of the bioactive hormone in the circulation. Furthermore, it remains to be established how PFCs penetrate cells and barriers and what are the mechanisms of clearance. In conclusion, we present both clinical and experimental evidence supporting the endocrine disrupting activity of PFCs on androgenic function, which is mediated by the AR. The interference of PFCs on the binding and activation of T on the AR, could explain the resulting alterations of seminal parameters and the reduction in testicular volume and penile length, together with shorter AGD, observed in young males from an exposure area. At the hormonal level, the reduced activation of T results in increased serum T levels, possibly due to the positive feedback on the hypothalamic-pituitary axis, as reflected by increased LH. Importantly, the antagonistic activity on T by PFCs could also extend to other steroids, such as DHT, progesterone or estradiol thereby affecting early and late development of male genital tract to the different extents. The Journal of Clinical Endocrinology & Metabolism; Copyright 2018 DOI: 10.1210/jc.2018-01855 *Corresponding author and person to whom reprint requests should be addressed: Prof. Carlo Foresta, Department of Medicine, Unit of Andrology and Medicine of Human Reproduction, University of Padova, Via Giustiniani, 2, 35128 Padova, Italy. Phone: +39-049-8218517; Fax: +39-049-8218520; e-mail: carlo.foresta@unipd.it D V A N C E A LE R T IC References 1. Conder JM, Hoke RA, De Wolf W, Russell MH, Buck RC. Are PFCAs bioaccumulative? A critical review and comparison with regulatory criteria and persistent lipophilic compounds. Environ. Sci. Technol. 2008;42(4):995–1003. 2. Steenland K, Fletcher T, Savitz DA. Epidemiologic evidence on the health effects of perfluorooctanoic acid (PFOA). Environ. Health Perspect. 2010;118(8):1100–1108. 3. Austin ME, Kasturi BS, Barber M, Kannan K, MohanKumar PS, MohanKumar SMJ. Neuroendocrine effects of perfluorooctane sulfonate in rats. Environ. Health Perspect. 2003;111(12):1485–9. 4. Inoue K, Okada F, Ito R, Kato S, Sasaki S, Nakajima S, Uno A, Saijo Y, Sata F, Yoshimura Y, Kishi R, Nakazawa H. Perfluorooctane sulfonate (PFOS) and related perfluorinated compounds in human maternal and cord blood samples: assessment of PFOS exposure in a susceptible population during pregnancy. Environ. Health Perspect. 2004;112(11):1204–7. 5. Fei C, McLaughlin JK, Tarone RE, Olsen J. Perfluorinated chemicals and fetal growth: a study within the Danish National Birth Cohort. Environ. Health Perspect. 2007;115(11):1677–82. 6. Kim S, Choi K, Ji K, Seo J, Kho Y, Park J, Kim S, Park S, Hwang I, Jeon J, Yang H, Giesy JP. Trans-Placental Transfer of Thirteen Perfluorinated Compounds and Relations with Fetal Thyroid Hormones. Environ. Sci. Technol. 2011;45(17):7465–7472. 7. Li N, Mruk DD, Chen H, Wong CKC, Lee WM, Cheng CY. Rescue of perfluorooctanesulfonate (PFOS)-mediated Sertoli cell injury by overexpression of gap junction protein connexin 43. Sci. Rep. 2016;6(1):29667. 8. Kim SJ, Heo SH, Lee DS, Hwang IG, Lee YB, Cho HY. Gender differences in pharmacokinetics and tissue distribution of 3 perfluoroalkyl and polyfluoroalkyl substances in rats. Food Chem. Toxicol. 2016;97:243–255. 9. Lau C, Anitole K, Hodes C, Lai D, Pfahles-Hutchens A, Seed J. Perfluoroalkyl acids: a review of monitoring and toxicological findings. Toxicol. Sci. 2007;99(2):366–94. 10. Kudo N, Kawashima Y. Toxicity and toxicokinetics of perfluorooctanoic acid in humans and animals. J. Toxicol. Sci. 2003;28(2):49–57. 11. Kennedy GL, Butenhoff JL, Olsen GW, O’Connor JC, Seacat AM, Perkins RG, Biegel LB, Murphy SR, Farrar DG. The toxicology of perfluorooctanoate. Crit. Rev. Toxicol. 34(4):351–84. 12. Foresta C, Tescari S, Di Nisio A. Impact of perfluorochemicals on human health and reproduction: a male’s perspective. J. Endocrinol. Invest. 2018;41(6):639–645. A ADVANCE ARTICLE: JCEM THE JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM Disclosure statement: The authors have declared that no conflict of interest exists. 10 Downloaded from https://academic.oup.com/jcem/advance-article-abstract/doi/10.1210/jc.2018-01855/5158211 by Endocrine Society Main Account user on 26 November 2018 A.D.N., I.S. and C.F. designed the study; U.V. and A.G. conducted clinical evaluations; A.D.N., I.S., M.S.R. performed molecular experiments, M.P., S.T., S.D.A. and D.G. conducted analytical experiments; L.A. and N.P. conducted biochemical experiments; A.D.N. , A.G. and C.F. participated to data analyses; A.D.N. and C.F. wrote the manuscript and all authors approved the final version. The Journal of Clinical Endocrinology & Metabolism; Copyright 2018 DOI: 10.1210/jc.2018-01855 LE IC T R A E C N A V D THE JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM ADVANCE ARTICLE: JCEM A 11 Downloaded from https://academic.oup.com/jcem/advance-article-abstract/doi/10.1210/jc.2018-01855/5158211 by Endocrine Society Main Account user on 26 November 2018 13. Skakkebaek NE, Rajpert-De Meyts E, Main KM. Testicular dysgenesis syndrome: an increasingly common developmental disorder with environmental aspects. Hum. Reprod. 2001;16(5):972–8. 14. Acerini CL, Miles HL, Dunger DB, Ong KK, Hughes IA. The descriptive epidemiology of congenital and acquired cryptorchidism in a UK infant cohort. Arch. Dis. Child. 2009;94(11):868–72. 15. Hauser R, Meeker JD, Duty S, Silva MJ, Calafat AM. Altered semen quality in relation to urinary concentrations of phthalate monoester and oxidative metabolites. Epidemiology 2006;17(6):682–91. 16. Vested A, Ramlau-Hansen CH, Olsen SF, Bonde JP, Kristensen SL, Halldorsson TI, Becher G, Haug LS, Ernst EH, 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(4):453–458. 17. Lau C. Perfluorinated Compounds. In: EXS.Vol 101.; 2012:47–86. 18. Kjeldsen LS, Bonefeld-Jørgensen EC. Perfluorinated compounds affect the function of sex hormone receptors. Environ. Sci. Pollut. Res. 2013;20(11):8031–8044. 19. 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. 20. 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(1):229–40. 21. Jensen AA, Leffers H. Emerging endocrine disrupters: perfluoroalkylated substances. Int. J. Androl. 2008;31(2):161–169. 22. Di Nisio A, Foresta C. Supplemental Figure 1. 2018. Available at: https://figshare.com/articles/Supplemental_Figure_1/7016234. 23. Veneto Region. D.G.R. 2133/2016, Annex A and subsequent modifications. 2016. 24. Petak SM, Nankin HR, Spark RF, Swerdloff RS, Rodriguez-Rigau LJ, American Association of Clinical Endocrinologists. American Association of Clinical Endocrinologists Medical Guidelines for clinical practice for the evaluation and treatment of hypogonadism in adult male patients--2002 update. Endocr. Pract. 2002;8(6):440–56. 25. Cameron N. Standards for human growth--their construction and use. S. Afr. Med. J. 1986;70(7):422–5. 26. Cacciari E, Milani S, Balsamo A, Dammacco F, De Luca F, Chiarelli F, Pasquino A, Tonini G, Vanelli M. Italian cross-sectional growth charts for height, weight and BMI (6–20 y). Eur. J. Clin. Nutr. 2002;56(2):171–180. 27. World Health Organization. Obesity: preventing and managing the global epidemic. Technical Report Series no. 894. (World Health Organization, ed.). Geneve, Switzerland: WHO Press; 2000. 28. Steele MF, Mattox JW. Correlation of arm-span and height in young women of two races. Ann. Hum. Biol. 14(5):445–7. 29. Zinn SL. Body Size and Habitus. In: Walker HK, Hall WD, Hurst JW (eds). Clinical methods: the history, physical, and laboratory examinations. 3rd ed. Boston: Butterworths. 1990, Chapter 137. 30. Ponchietti R, Mondaini N, Bonafè M, Di Loro F, Biscioni S, Masieri L. Penile length and circumference: a study on 3,300 young Italian males. Eur. Urol. 2001;39(2):183–6. 31. Parra MD, Mendiola J, Jørgensen N, Swan SH, Torres-Cantero AM. Anogenital distance and reproductive parameters in young men. Andrologia 2016;48(1):3–10. 32. World Health Organization. WHO Laboratory Manual for the Examination and Processing of Human Semen. (World Health Organization, ed.). Geneve, Switzerland: WHO Press; 2010. The Journal of Clinical Endocrinology & Metabolism; Copyright 2018 DOI: 10.1210/jc.2018-01855 LE IC T R A E C N A V D THE JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM ADVANCE ARTICLE: JCEM A 12 Downloaded from https://academic.oup.com/jcem/advance-article-abstract/doi/10.1210/jc.2018-01855/5158211 by Endocrine Society Main Account user on 26 November 2018 33. Zuccarello D, Ferlin A, Vinanzi C, Prana E, Garolla A, Callewaert L, Claessens F, Brinkmann AO, Foresta C. Detailed functional studies on androgen receptor mild mutations demonstrate their association with male infertility. Clin. Endocrinol. (Oxf). 2008;68(4):580– 588. 34. Ascoli M. Regulation of gonadotropin receptors and gonadotropin responses in a clonal strain of Leydig tumor cells by epidermal growth factor. J. Biol. Chem. 1981;256(1):179–83. 35. Ingelido AM, Abballe A, Gemma S, Dellatte E, Iacovella N, De Angelis G, Zampaglioni F, Marra V, Miniero R, Valentini S, Russo F, Vazzoler M, Testai E, De Felip E. Biomonitoring of perfluorinated compounds in adults exposed to contaminated drinking water in the Veneto Region, Italy. Environ. Int. 2018;110:149–159. 36. Ingelido AM, Marra V, Abballe A, Valentini S, Iacovella N, Barbieri P, Porpora MG, Domenico A di, Felip E De. Perfluorooctanesulfonate and perfluorooctanoic acid exposures of the Italian general population. Chemosphere 2010;80(10):1125–1130. 37. Ministero dell’Ambiente e della Tutela del Territorio e del Mare e Istituto di Ricerca sulle Acque. Realizzazione di uno studio di valutazione del Rischio Ambientale e Sanitario associato alla contaminazione da sostanze perfluoro-alchiliche (PFAS) nel Bacino del Po e nei principali bacini fluviali italiani (in Italian). 2010. Available at: http://www.minambiente.it/sites/default/files/archivio/allegati/reach/progettoPFAS_ottobre20 13.pdf. 38. Veneto Region. Press release N° 961 (in Italian) of 11/07/2018. 2018. Available at: https://www.regione.veneto.it/web/guest/comunicati-stampa/dettagliocomunicati?_spp_detailId=3220944. 39. Mitchell RT, Mungall W, McKinnell C, Sharpe RM, Cruickshanks L, Milne L, Smith LB. Anogenital distance plasticity in adulthood: implications for its use as a biomarker of fetal androgen action. Endocrinology 2015;156(1):24–31. 40. Adibi JJ, Lee MK, Naimi AI, Barrett E, Nguyen RH, Sathyanarayana S, Zhao Y, Thiet M-P, Redmon JB, Swan SH. Human Chorionic Gonadotropin Partially Mediates Phthalate Association With Male and Female Anogenital Distance. J. Clin. Endocrinol. Metab. 2015;100(9):E1216-24. 41. Vafeiadi M, Agramunt S, Papadopoulou E, Besselink H, Mathianaki K, Karakosta P, Spanaki A, Koutis A, Chatzi L, Vrijheid M, Kogevinas M. In Utero Exposure to Dioxins and Dioxin-like Compounds and Anogenital Distance in Newborns and Infants. Environ. Health Perspect. 2012;121(1):125–130. 42. Miao M, Yuan W, He Y, Zhou Z, Wang J, Gao E, Li G, Li D-K. In utero exposure to bisphenol-A and anogenital distance of male offspring. Birth Defects Res. A. Clin. Mol. Teratol. 2011;91(10):867–72. 43. ARPAV (The Regional Agency of Environmental Protection of Veneto Region). Stato dell’inquinamento da sostanze perfluoroalchilice (PFAS) in provincial di Vicenza, Padova, Verona (in Italian). 2013. Available at: http://www.arpa.veneto.it/temiambientali/acqua/file-e-allegati/documenti/acque-interne/pfas/Nota Tecnica PFAS.pdf. 44. Biegel LB, Liu RCM, Hurtt ME, Cook JC. Effects of Ammonium Perfluorooctanoate on Leydig-Cell Function: In Vitro, in Vivo, and ex Vivo Studies. Toxicol. Appl. Pharmacol. 1995;134(1):18–25. 45. Shi Z, Zhang H, Liu Y, Xu M, Dai J. Alterations in Gene Expression and Testosterone Synthesis in the Testes of Male Rats Exposed to Perfluorododecanoic Acid. Toxicol. Sci. 2007;98(1):206–215. 46. Wan HT, Zhao YG, Wong MH, Lee KF, Yeung WSB, Giesy JP, Wong CKC. Testicular Signaling Is the Potential Target of Perfluorooctanesulfonate-Mediated Subfertility in Male Mice1. Biol. Reprod. 2011;84(5):1016–1023. The Journal of Clinical Endocrinology & Metabolism; Copyright 2018 DOI: 10.1210/jc.2018-01855 LE IC T R A E C N A V D A THE JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM ADVANCE ARTICLE: JCEM Figure 1. Total testosterone levels are positively correlated with serum PFOA. Correlation between serum log-transformed PFOA levels and total Testosterone in the exposed group (full circles, straight line, n =50) and in control group (empty circles, dotted line, n =50). Figure 2. PFOA and PFOS inhibit androgen receptor transactivation in HeLa transfected cells. AR gene reporter assay on HeLa cells transfected with Luc-AR and stimulated with T 10 nM and PFOA 1 uM or PFOS 1 uM, alone or in combination with T. Flutamide (Flut 1uM) served as internal negative control of inhibitory activity on AR. AR activity is reported as relative (%) to positive control (T 10 nM, set to 100%). Data are 13 Downloaded from https://academic.oup.com/jcem/advance-article-abstract/doi/10.1210/jc.2018-01855/5158211 by Endocrine Society Main Account user on 26 November 2018 47. 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(7):3370–3385. 48. Kang JS, Choi JS, Park JW. Transcriptional changes in steroidogenesis by perfluoroalkyl acids (PFOA and PFOS) regulate the synthesis of sex hormones in H295R cells. Chemosphere 2016;155:436–443. 49. Joensen UN, Bossi R, Leffers H, Jensen AA, Skakkebæk NE, Jørgensen N. Do Perfluoroalkyl compounds impair human semen quality? Environ. Health Perspect. 2009;117(6):923–927. 50. Toft G, Jönsson BAG, Lindh CH, Giwercman A, Spano M, Heederik D, Lenters V, Vermeulen R, Rylander L, Pedersen HS, Ludwicki JK, Zviezdai V, Bonde JP. Exposure to perfluorinated compounds and human semen quality in arctic and European populations. Hum. Reprod. 2012;27(8):2532–2540. 51. Singh RP, Shafeeque CM, Sharma SK, Pandey NK, Singh R, Mohan J, Kolluri G, Saxena M, Sharma B, Sastry KVH, Kataria JM, Azeez PA. Bisphenol A reduces fertilizing ability and motility by compromising mitochondrial function of sperm. Environ. Toxicol. Chem. 2015;34(7):1617–1622. 52. Raymer JH, Michael LC, Studabaker WB, Olsen GW, Sloan CS, Wilcosky T, Walmer DK. Concentrations of perfluorooctane sulfonate (PFOS) and perfluorooctanoate (PFOA) and their associations with human semen quality measurements. Reprod. Toxicol. 2012;33(4):419–427. 53. Eriksen KT, Sorensen M, McLaughlin JK, Lipworth L, Tjonneland A, Overvad K, Raaschou-Nielsen O. Perfluorooctanoate and perfluorooctanesulfonate plasma levels and risk of cancer in the general danish population. J. Natl. Cancer Inst. 2009;101(8):605–609. 54. Behr A-C, Lichtenstein D, Braeuning A, Lampen A, Buhrke T. Perfluoroalkylated substances (PFAS) affect neither estrogen and androgen receptor activity nor steroidogenesis in human cells in vitro. Toxicol. Lett. 2018;291:51–60. 55. Du G, Hu J, Huang H, Qin Y, Han X, Wu D, Song L, Xia Y, Wang X. Perfluorooctane sulfonate (PFOS) affects hormone receptor activity, steroidogenesis, and expression of endocrine-related genes in vitro and in vivo. Environ. Toxicol. Chem. 2013;32(2):353–60. 56. Hölzer J, Midasch O, Rauchfuss K, Kraft M, Reupert R, Angerer J, Kleeschulte P, Marschall N, Wilhelm M. Biomonitoring of perfluorinated compounds in children and adults exposed to perfluorooctanoate-contaminated drinking water. Environ. Health Perspect. 2008;116(5):651–7. 57. Olsen GW, Logan PW, Hansen KJ, Simpson CA, Burris JM, Burlew MM, Vorarath PP, Venkateswarlu P, Schumpert JC, Mandel JH. An occupational exposure assessment of a perfluorooctanesulfonyl fluoride production site: biomonitoring. AIHA J. (Fairfax,. Va). 2003;64(5):651–9. The Journal of Clinical Endocrinology & Metabolism; Copyright 2018 DOI: 10.1210/jc.2018-01855 IC T A R Figure 4. Expression of androgen receptor in murine Leydig MA-10 cells under different conditions. Immunofluorescence of AR (FITC, green) nuclear translocation in Leydig Ma-10 cells stimulated with T and PFOA at different concentrations as reported in the figure. Flutamide (1 uM) served as internal control. Nuclei are stained with DAPI (blue). Cells were visualized by scanning confocal laser microscopy (60x magnifcation). V A N C E Figure 5. Androgen receptor nuclear translocation induced by testosterone is reduced by PFOA in murine MA-10 Leydig cells. Relative quantification (%) of nuclear fluorescence intensity with respect to total fluoresce intensity in MA-10 Leydig cells stimulated with T and PFOA at different concentrations. Results are the mean ± SE. The Kruskal-Wallis test was used to compare differences between concentrations. The Mann– Whitney test with Bonferroni correction was used to compare each concentration with the control. The same procedure was applied to comparisons between different stimuli (PFOA 0.1, 1 and 10 µM and flutamide 1 µM) and T, within each T concentration. *= p<0.05 vs T; **= p<0.001 vs T; †= p<0.05 vs T 0, 10 and 100 nM; ‡= p<0.05 vs T 0, 1 and 100 nM; §= p <0.05 vs T 0, 1 and 10 nM; ¥= p<0.05 vs T 0 nM; ç= p<0.05 vs 0 and 1 nM D Table 1. Anthropometric and seminal parameters in 171 controls and 212 exposed subjects. Parameters Age (yrs.) A ADVANCE ARTICLE: JCEM THE JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM LE Figure 3. Binding of testosterone to immobilized androgen receptor is reduced by PFOA. Solutions of testosterone (288.42 Da) (A) and PFOA (414.07 Da) (C) were injected at a flow rate of 30 µl/min at 25°C, using 10mM Hepes pH7.4, 0.15M NaCl containing 3% MeOH (V/V) as running buffer. Each SPR trace was subtracted for unspecific binding (<2% of RUmax). The response units (RU) at the steady state were plotted as a function of [analyte] and fitted to the Langmuir equation to yield the dissociation constant Kd (B, D). No interaction between PFOA (up to 4mM) and AR was detected under the same experimental conditions. (E) Next, we performed competition experiment to assess whether the presence of PFOA would reduce the binding of testosterone to AR. A 250 mM solution of testosterone was incubated with different concentrations of PFOA (0-4 µM) for at least 10 minutes before injection over the same AR-coated sensor chip. We observed a ∼35% reduction of testosterone binding to AR at 4 µM PFOA. Results are shown as the maximal association response units (RU, expressed as the percentage relative to the response measured without PFOA) achieved at increasing concentrations of PFOA. (F) Raw data showing the inhibitory effect of PFOA at 1 µM (red) and the reproducibility of testosterone binding before and after the competition experiment (black and blue). Height (cm) Mea ± SD n 18.7 ± 1.0 179.2 ± 6.2 Weight (kg) 73.2 ± 8.5 BMI (kg/m²) 22.8 ± 2.3 WC (cm) 81.8 ± 7.1 Arm span 182.1 ± 10.2 (cm) Crown-topubis 81.8 ± 4.9 distance (cm) Pubis-to97.4 ± 5.3 floor Controls (N=171)* Media min-max n 18.0-24.0 18.0 162.0180.0 192.0 54.0-96.0 73.0 18.2-31.0 22.5 64.0-103.0 81.0 87.0-200.0 (IQR) (18.0-19.0) (175.0184.0) (67.0-78.0) (21.1-24.0) (77.0-85.6) (178.0182.0 187.6) Exposed (N=212)* Media min-max n 18.0-22.0 18.0 160.0178.8 ± 6.9 179.0 203.0 73.9 ± 11.9 47.0-120.0 73.0 23.1 ± 3.1 16.6-35.8 22.5 84.0 ± 10.5 63.5-140.0 82.0 160.0182.0 ± 8.2 181.5 204.0 Mea ± SD n 18.5 ± 0.8 (IQR) (18.0-19.0) (175.0183.0) (65.0-80.0) (21.0-24.5) (77.0-88.0) (176.5187.0) Raw Adj. pa pb 0.081 0.567 0.575 1.0 0.897 0.492 0.174 1.0 1.0 0.87 0.276 1.0 70.0-94.0 82.0 (78.0-85.2) 82.9 ± 5.5 68.0-98.0 83.0 (79.0-86.0) 0.041 0.328 84.5-110.0 97.8 (93.0-101.2) 95.9 ± 5.7 79.0-117.0 96.0 (93.0-100.0) 0.009 0.09 14 Downloaded from https://academic.oup.com/jcem/advance-article-abstract/doi/10.1210/jc.2018-01855/5158211 by Endocrine Society Main Account user on 26 November 2018 reported as mean±SD of three independent experiments. *p <0.001 calculated with one-way ANOVA with a Dunnett´s post hoc test with T 10 nM as reference category. 0.68-1.01 0.8 (0.8-0.9) 0.9 ± 0.1 0.6-1.2 0.9 (0.8-0.9) 7.6-26.5 16.0 (14.1-18.0) 14.7 ± 3.2 6.8-24.5 14.5 (12.5-16.5) <0.001 <0.001 6.0-13.5 10.0 (8.5-11.0) 8.6 ± 1.7 2.0-13.5 9.0 (8.0-10.0) <0.001 <0.001 5.0-13.0 10.0 (9.5-10.5) 9.9 ± 1.1 7.0-13.0 10.0 (9.0-10.5) 2.5-7.2 4.5 (4.0-5.0) 4.1 ± 0.9 2.0-7.0 4.0 (3.5-4.5) 0.3-7.5 2.5 (1.5-3.5) 2.6 ± 1.3 0.2-7.0 2.5 (1.5-3.3) 7.0-8.0 7.5 (7.4-7.6) 7.6 ± 0.2 7.0-8.5 7.7 (7.5-7.7) 66.2 ± 53.2 0-327.0 57.0 (25.3-99.0) 0.045 0.180 154. 5 0-817.5 123.0 (43.4-258.0) 0.032 0.160 135.0 (66.0-281.1) 166.8 ± 53.0 (42.0-62.0) 44.1 ± 17.1 0-85.0 44.0 (32.0-57.0) 0-32.0 6.0 (3.0-10.0) 8.2 ± 7.6 0-63.0 6.0 (4.0-10.0) 0-91.0 38.0 (30.0-50.3) 46.8 ± 17.0 0-30.0 6.0 (4.0-12.0) 0-98.0 85.0 (78.0-90.0) T 0-91.0 R 0-90.0 0.134 0.126 0.804 <0.001 <0.001 0.512 0.568 LE 0-2240 65.0 (33.2-115.6) IC 0-800.0 0.014 47.0 (35.0-57.0) <0.001 <0.001 <0.001 <0.001 0.284 0.568 <0.001 <0.001 6.1 ± 4.3 0-20.0 6.0 (2.0-8.0) 0.006 0.036 81.1 ± 9.7 0-95.0 83.0 (78.0-88.0) 0.057 0.180 E A IQR: interquartile range 25th – 75th percentiles. Significant p values are in bold. *In the seminal parameters analyses, 1 subject within each group failed to collect semen and was therefore omitted. aMann-Whitney test was used to assess differences between groups. bAdjustment for multiple comparisons was calculated with the Bonferroni-Holm method. C Table 2. Sex hormones, PFOA and PFOS levels in serum and semen from 50 controls and 50 exposed subjects, with respective anthropometrics and seminal parameters. Parameter 4.71 ± 2.08 1.2-8.0 4.70 (3.5-6.6) 0.89 ± 0.7 0.6-1.8 ± 4.06 6.8-29.4 ± 1.26 ± 1.63 1.2-6.6 1.4-8.3 18.233.6 64.0114.0 165.0200.0 71.088.5 87.0106.0 0.82 (0.4-1.3) (0.080.1 0.11) (0.08-00.11 13) (12.918.98 17.9) 2.89 (2.0-3.8) 4.18 (2.9-6.8) (21.122.19 24.7) (77.080.75 86.0) 182.7 (178.05 185.8) (79.082.0 85.0) (93.097.0 101.1) 0.7-1.0 0.85 (0.8-0.9) A Serum PFOA (ng/ml) Serum PFOS (ng/ml) 0.1 ± 0.01 0.0-0.1 0.11 ± 0.03 0.1-0.2 V Semen PFOA (ng/ml) Semen PFOS (ng/ml) D Testosterone (nmol/L) FSH (U/L) LH(U/L) BMI (kg/m²) WC (cm) Arm span (cm) Crown-to-pubis distance (cm) Pubis-to-floor distance (cm) Crown-to-pubis/pubis-tofloor ratio Controls (N =50) minMedi max an (IQR) N Mea n ± SD A ADVANCE ARTICLE: JCEM distance (cm) Crown-topubis/pubis0.8 ± 0.1 to-floor ratio Testicular 16.1 ± 3.2 Volume (ml) Penis length 9.7 ± 1.6 (cm) Penis circumferenc 10.0 ± 1.0 e (cm) AGD (cm) 4.5 ± 0.8 Semen 2.7 ± 1.4 volume (mL) pH 7.5 ± 0.2 Sperm concentratio 89.2 ± 97.9 n (10⁶/mL) Total sperm 292. 230.5 ± 6 count (10⁶) Progressive 51.8 ± 15.5 motility (%) Non progressive 7.5 ± 6.5 motility (%) Immotile 40.2 ± 14.2 sperm (%) Normal morphology 7.9 ± 5.8 (%) Viability (%) 82.4 ± 10.0 DOI: 10.1210/jc.2018-01855 15.4 2 3.03 4.24 23.0 2 82.4 8 182. 19 81.5 3 96.8 0 ± 2.86 ± 8.42 ± 6.61 ± 4.17 ± 5.20 0.84 ± 0.06 Testicular Volume (ml) 16.8 ± 3.61 6 9.7-26.5 Penis length (cm) 10.0 ± 1.87 6.0-13.0 (14.816.13 19.0) 10.0 (9.0-11.0) 15 Exposed (N =50) minMedi max an (IQR) 2.37.35 (4.7-14.9) 156.7 0.0-4.0 1.11 (0.8-1.3) (0.110.0-5.3 0.24 0.99) (0.010.06 0.0-1.1 0.11 0.14) (16.35.27 9.3-35.0 18.98 21.8) 1.53 1.5-7.3 2.99 (2.2-7.0) 1.79 2.0-8.4 5.37 (4.3-7.0) 16.6(20.43.24 22.40 30.5 25.5) 11.5 65.0(76.581.50 2 140.0 85.4) 160.0179.0 (174.27.47 198.0 0 187.0) 74.0(79.04.92 82.0 96.0 84.0) 84.0(90.35.27 95.0 102.0 99.0) 0.730.08 0.86 (0.8-0.9) 1.14 (12.63.32 9.5-24.5 14.00 16.0) Mea n ± SD 14.9 25.0 ± 9 8 1.11 ± 0.3 0.90 0.67 ± 8 Raw Adj. pa pb <0.00 <0.00 1 1 0.012 0.048 <0.00 <0.00 1 1 0.12 ± 0.916 0.916 <0.00 <0.00 1 1 0.228 0.576 0.003 0.015 19.3 4 3.48 5.47 22.9 0 83.1 3 180. 45 82.1 4 94.5 9 ± ± ± ± ± ± ± ± 0.87 ± 14.6 ± 7 8.75 ± 1.82 4.0-12.0 9.00 (8.0-10.0) 0.847 1.000 0.757 1.000 0.246 0.738 0.592 1.000 0.064 0.320 0.110 0.440 <0.00 <0.00 1 1 <0.00 <0.00 1 1 Downloaded from https://academic.oup.com/jcem/advance-article-abstract/doi/10.1210/jc.2018-01855/5158211 by Endocrine Society Main Account user on 26 November 2018 THE JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM The Journal of Clinical Endocrinology & Metabolism; Copyright 2018 Penis circumference (cm) Total sperm count (10⁶) Progressive motility (%) ± 0.90 9.0-13.0 10.10 (9.9-11.0) 9.65 ± 0.90 7.8-12.0 ± 0.9 ± 1.91 ± 0.21 133. ± 87 347. ± 40 13.3 ± 9 2.7-7.2 0.5-9.0 7.0-7.9 6.0800.0 4.2 2.76 7.62 89.7 6 226. 93 55.3 1 2.5-5.7 0.3-6.0 7.0-8.0 6.0264.0 30.085.0 4.50 (4.0-5.2) 2.55 (1.5-3.9) 7.60 (7.5-7.7) (27.449.50 94.3) 146.2 (70.05 270.3) (45.054.50 64.8) 4.50 (3.0-9.8) Non progressive motility (%) 6.88 ± 6.38 0.0-27.0 Immotile sperm (%) 39.0 12.3 ± 8 4 15.068.0 Normal morphology (%) 8.72 ± 5.51 2.0-20.0 Viability (%) 82.4 ± 7.81 4 53.092.0 38.0 (30.246.5) 15.8-680 15.088.0 5.06 ± 3.35 1.0-21.0 39.6 16.4 ± 3 9 7.0-79.0 7.0 (4.0-12.0) 4.55 ± 2.31 2.0-10.0 (80.089.5) 79.6 ± 7.67 9 60.091.0 82.50 th a 4.00 (3.5-5.0) 3.00 (1.6-3.4) 7.70 (7.6-7.7) (34.454.50 96.5) 171.5 (77.25 301.7) (43.057.0 68.0) 4.0 (3.0-6.0) 40.50 (25.553.5) <0.00 1 0.019 0.373 0.005 <0.00 1 0.114 1.000 0.042 0.771 1.000 0.596 1.000 0.992 1.000 0.106 0.636 0.624 1.000 4.0 (2.0-6.0) 82.0 (75.085.0) <0.00 <0.00 1 1 0.048 0.336 D V A N C E A R T IC IQR: interquartile range 25 – 75 percentiles. Significant p values are in bold. Mann-Whitney test was used to assess differences between groups. bAdjustment for multiple comparisons was calculated with the BonferroniHolm method. A THE JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM th ADVANCE ARTICLE: JCEM 6.1-2240 ± 0.8 ± 1.37 ± 0.23 59.3 ± 5 264. ± 38 16.4 ± 1 9.50 (9.0-10.0) 16 Downloaded from https://academic.oup.com/jcem/advance-article-abstract/doi/10.1210/jc.2018-01855/5158211 by Endocrine Society Main Account user on 26 November 2018 AGD (cm) Semen volume (mL) pH Sperm concentration (10⁶/mL) 10.3 1 4.5 3.09 7.55 92.4 0 241. 78 54.7 3 DOI: 10.1210/jc.2018-01855 LE The Journal of Clinical Endocrinology & Metabolism; Copyright 2018 V D E C N IC LE T R A THE JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM ADVANCE ARTICLE: ADVANCE ARTICLE: Endocrinology JCEM A A Downloaded from https://academic.oup.com/jcem/advance-article-abstract/doi/10.1210/jc.2018-01855/5158211 by Endocrine Society Main Account user on 26 November 2018 V D E C N IC LE T R A THE JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM ADVANCE ARTICLE: ADVANCE ARTICLE: Endocrinology JCEM A A Downloaded from https://academic.oup.com/jcem/advance-article-abstract/doi/10.1210/jc.2018-01855/5158211 by Endocrine Society Main Account user on 26 November 2018 V D E C N IC LE T R A THE JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM ADVANCE ARTICLE: ADVANCE ARTICLE: Endocrinology JCEM A A Downloaded from https://academic.oup.com/jcem/advance-article-abstract/doi/10.1210/jc.2018-01855/5158211 by Endocrine Society Main Account user on 26 November 2018 V D E C N IC LE T R A THE JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM ADVANCE ARTICLE: ADVANCE ARTICLE: Endocrinology JCEM A A Downloaded from https://academic.oup.com/jcem/advance-article-abstract/doi/10.1210/jc.2018-01855/5158211 by Endocrine Society Main Account user on 26 November 2018 V D E C N IC LE T R A THE JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM ADVANCE ARTICLE: ADVANCE ARTICLE: Endocrinology JCEM A A Downloaded from https://academic.oup.com/jcem/advance-article-abstract/doi/10.1210/jc.2018-01855/5158211 by Endocrine Society Main Account user on 26 November 2018