Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 509
Filter
1.
Anal Chim Acta ; 1313: 342789, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-38862206

ABSTRACT

BACKGROUND: Therapeutic drug monitoring of treatment with therapeutic antibodies is hampered by the application of a wide range of different methods in the quantification of serum levels. LC-MS based methods could significantly improve comparability of results from different laboratories, but such methods are often considered complicated and costly. We developed a method for LC-MS/MS based quantification of 11 therapeutic antibodies concomitantly measured in a single run, with emphasis on simplicity in sample preparation and low cost. RESULTS: After a single-step sample purification using caprylic acid precipitation to remove interfering proteins, the sample underwent proteolysis followed by LC-MS/MS analysis. Infliximab is used as internal standard for sample preparation while isotope-labeled signature peptides identified for each analyte are internal standards for the LC-MS/MS normalization. The method was validated according to recognized guidelines, and pipetting steps can be performed by automated liquid handling systems. The total precision of the method ranged between 2.7 and 7.3 % (5.1 % average) across the quantification range of 4-256 µg/ml for all 11 drugs, with an average accuracy of 96.3 %. Matrix effects were xamined in 55 individual patient samples instead of the recommended 6, and 147 individual patient samples were screened for interfering compounds. SIGNIFICANCE AND NOVELTY: Our method for simultaneous quantification of 11 t-mAb in human serum allows an unprecedented integration of robustness, speed and reduced complexity, which could pave the way for uniform use in research projects and clinical settings alike. In addition, the first LC-MS protocol for signature peptide-based quantification of durvalumab is described. This high throughput method can be readily adapted to a drug panel of choice.


Subject(s)
Caprylates , Tandem Mass Spectrometry , Tandem Mass Spectrometry/methods , Tandem Mass Spectrometry/economics , Humans , Caprylates/chemistry , Caprylates/blood , Chemical Precipitation , Chromatography, Liquid/methods , High-Throughput Screening Assays/economics , Antibodies, Monoclonal/blood , Antibodies, Monoclonal/chemistry , Liquid Chromatography-Mass Spectrometry
2.
Environ Int ; 187: 108727, 2024 May.
Article in English | MEDLINE | ID: mdl-38735074

ABSTRACT

BACKGROUND: There is inconclusive evidence for an association between per- and polyfluoroalkyl substances (PFAS) and fetal growth. OBJECTIVES: We conducted a nation-wide register-based cohort study to assess the associations of the estimated maternal exposure to the sum (PFAS4) of perfluorooctane sulfonic acid (PFOS), perfluorooctanoic acid (PFOA), perfluorononanoic acid (PFNA) and perfluorohexane sulfonic acid (PFHxS) with birthweight as well as risk of small- (SGA) and large-for-gestational-age (LGA). MATERIALS AND METHODS: We included all births in Sweden during 2012-2018 of mothers residing ≥ four years prior to partus in localities served by municipal drinking water where PFAS were measured in raw and drinking water. Using a one-compartment toxicokinetic model we estimated cumulative maternal blood levels of PFAS4 during pregnancy by linking residential history, municipal PFAS water concentration and year-specific background serum PFAS concentrations in Sweden. Individual birth outcomes and covariates were obtained via register linkage. Mean values and 95 % confidence intervals (CI) of ß coefficients and odds ratios (OR) were estimated by linear and logistic regressions, respectively. Quantile g-computation regression was conducted to assess the impact of PFAS4 mixture. RESULTS: Among the 248,804 singleton newborns included, no overall association was observed for PFAS4 and birthweight or SGA. However, an association was seen for LGA, multivariable-adjusted OR 1.08 (95% CI: 1.01-1.16) when comparing the highest PFAS4 quartile to the lowest. These associations remained for mixture effect approach where all PFAS, except for PFOA, contributed with a positive weight. DISCUSSIONS: We observed an association of the sum of PFAS4 - especially PFOS - with increased risk of LGA, but not with SGA or birthweight. The limitations linked to the exposure assessment still require caution in the interpretation.


Subject(s)
Alkanesulfonic Acids , Birth Weight , Caprylates , Drinking Water , Fetal Development , Fluorocarbons , Maternal Exposure , Water Pollutants, Chemical , Fluorocarbons/blood , Fluorocarbons/analysis , Humans , Drinking Water/chemistry , Female , Sweden , Water Pollutants, Chemical/analysis , Water Pollutants, Chemical/blood , Pregnancy , Adult , Alkanesulfonic Acids/blood , Maternal Exposure/statistics & numerical data , Fetal Development/drug effects , Birth Weight/drug effects , Caprylates/blood , Infant, Newborn , Cohort Studies , Sulfonic Acids/blood , Registries , Male , Infant, Small for Gestational Age , Young Adult
3.
Int J Hyg Environ Health ; 259: 114387, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38703464

ABSTRACT

BACKGROUND: In the past, perfluorooctanoic acid (PFOA) was produced and applied as an emulsifier in a fluoropolymer production plant in the Altötting district, southern Bavaria (Germany). This chemical was released directly into the environment, resulting in the contamination of the local drinking water. During a human biomonitoring (HBM) survey in 2018, increased median PFOA blood serum levels, compared to a normally exposed control group with no known source of PFOA exposure from Munich, Germany, were detected in the resident population (23.18 µg/l in the general population, 20.71 µg/l in the children's group). The follow-up study aimed to investigate whether purification of the drinking water as the main PFOA exposure source has been successful in reducing internal PFOA exposure and to estimate the association of internal PFOA exposure with possible influencing factors. METHODS: Only individuals who had already participated in the HBM study in 2018 were included. For the determination of the PFOA serum concentration, 5 ml of blood was drawn from each participating person. Blood samples were collected in the period from June to August 2022. Furthermore, information on sociodemographic characteristics, health status, dietary behaviour and other lifestyle factors were collected by means of a self-administered questionnaire. To examine the association of PFOA blood serum levels with possible influencing factors, such as age, gender and consumption of fish and game meat, a logistic regression model with a PFOA value > 10 µg/l as outcome was used. RESULTS: A total of 764 individuals participated in the follow-up study in 2022. Analyses were performed separately for the general population (n = 559), women of reproductive age (15-49 years old) (n = 120), and children under 12 years old (n = 30). Median PFOA blood levels have decreased by 56.9% in the general population, by 59.8% in the group of women of reproductive age and by 73.4% in the group of children under 12 years old. In the general population, a higher probability of a PFOA value > 10 µg/l was found for those aged 40-59 years (Odds ratio (OR) = 2.33 (95%CI: 1.23 to 4.43, p = 0.01) and those aged 60 years and older (OR = 5.32, 95%CI: 2.78 to 10.19, p < 0.001). CONCLUSIONS: In all study groups, the median PFOA serum levels decreased as expected after a half-life of four years, which confirms that contamination via drinking water has ceased. Furthermore, our study identified age as a significant predictor of internal PFOA exposure, while no influence was found for the consumption of fish and game meat. Further investigations are needed to quantify in a more detailed way the influence of dietary habits on PFOA exposure.


Subject(s)
Biological Monitoring , Caprylates , Environmental Exposure , Fluorocarbons , Humans , Caprylates/blood , Fluorocarbons/blood , Germany , Female , Male , Adult , Middle Aged , Child , Adolescent , Young Adult , Environmental Exposure/analysis , Child, Preschool , Aged , Follow-Up Studies , Water Pollutants, Chemical/blood , Water Pollutants, Chemical/analysis , Infant , Environmental Pollutants/blood , Drinking Water/chemistry , Chemical Industry
4.
Sci Total Environ ; 933: 173157, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38740209

ABSTRACT

Per- and polyfluoroalkyl substances (PFAS) are related to various adverse health outcomes, and food is a common source of PFAS exposure. Dietary sources of PFAS have not been adequately explored among U.S. pregnant individuals. We examined associations of dietary factors during pregnancy with PFAS concentrations in maternal plasma and human milk in the New Hampshire Birth Cohort Study. PFAS concentrations, including perfluorohexane sulfonate (PFHxS), perfluorooctane sulfonate (PFOS), perfluorooctanoate (PFOA), perfluorononanoate (PFNA), and perfluorodecanoate (PFDA), were measured in maternal plasma collected at ∼28 gestational weeks and human milk collected at ∼6 postpartum weeks. Sociodemographic, lifestyle and reproductive factors were collected from prenatal questionnaires and diet from food frequency questionnaires at ∼28 gestational weeks. We used adaptive elastic net (AENET) to identify important dietary variables for PFAS concentrations. We used multivariable linear regression to assess associations of dietary variables selected by AENET models with PFAS concentrations. Models were adjusted for sociodemographic, lifestyle, and reproductive factors, as well as gestational week of blood sample collection (plasma PFAS), postpartum week of milk sample collection (milk PFAS), and enrollment year. A higher intake of fish/seafood, eggs, coffee, or white rice during pregnancy was associated with higher plasma or milk PFAS concentrations. For example, every 1 standard deviation (SD) servings/day increase in egg intake during pregnancy was associated with 4.4 % (95 % CI: 0.6, 8.4), 3.3 % (0.1, 6.7), and 10.3 % (5.6, 15.2) higher plasma PFOS, PFOA, and PFDA concentrations respectively. Similarly, every 1 SD servings/day increase in white rice intake during pregnancy was associated with 7.5 % (95 % CI: -0.2, 15.8) and 12.4 % (4.8, 20.5) greater milk PFOS and PFOA concentrations, respectively. Our study suggests that certain dietary factors during pregnancy may contribute to higher PFAS concentrations in maternal plasma and human milk, which could inform interventions to reduce PFAS exposure for both birthing people and offspring.


Subject(s)
Alkanesulfonic Acids , Diet , Environmental Pollutants , Fluorocarbons , Milk, Human , Humans , Fluorocarbons/blood , Fluorocarbons/analysis , Milk, Human/chemistry , Female , Diet/statistics & numerical data , Environmental Pollutants/blood , Environmental Pollutants/analysis , New Hampshire , Alkanesulfonic Acids/analysis , Alkanesulfonic Acids/blood , Adult , Birth Cohort , Maternal Exposure/statistics & numerical data , Pregnancy , Caprylates/blood , Caprylates/analysis , Cohort Studies , Dietary Exposure/statistics & numerical data , Dietary Exposure/analysis , Decanoic Acids/blood , Decanoic Acids/analysis
5.
Environ Int ; 186: 108621, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38593693

ABSTRACT

In utero and children's exposure to per- and polyfluoroalkyl substances (PFAS) is a major concern in health risk assessment as early life exposures are suspected to induce adverse health effects. Our work aims to estimate children's exposure (from birth to 12 years old) to PFOA and PFOS, using a Physiologically-Based Pharmacokinetic (PBPK) modelling approach. A model for PFAS was updated to simulate the internal PFAS exposures during the in utero life and childhood, and including individual characteristics and exposure scenarios (e.g., duration of breastfeeding, weight at birth, etc.). Our approach was applied to the HELIX cohort, involving 1,239 mother-child pairs with measured PFOA and PFOS plasma concentrations at two sampling times: maternal and child plasma concentrations (6 to 12 y.o). Our model predicted an increase in plasma concentrations during fetal development and childhood until 2 y.o when the maximum concentrations were reached. Higher plasma concentrations of PFOA than PFOS were predicted until 2 y.o, and then PFOS concentrations gradually became higher than PFOA concentrations. From 2 to 8 y.o, mean concentrations decreased from 3.1 to 1.88 µg/L or ng/mL (PFOA) and from 4.77 to 3.56 µg/L (PFOS). The concentration-time profiles vary with the age and were mostly influenced by in utero exposure (on the first 4 months after birth), breastfeeding (from 5 months to 2 (PFOA) or 5 (PFOS) y.o of the children), and food intake (after 3 (PFOA) or 6 (PFOS) y.o of the children). Similar measured biomarker levels can correspond to large differences in the simulated internal exposures, highlighting the importance to investigate the children's exposure over the early life to improve exposure classification. Our approach demonstrates the possibility to simulate individual internal exposures using PBPK models when measured biomarkers are scarce, helping risk assessors in gaining insight into internal exposure during critical windows, such as early life.


Subject(s)
Alkanesulfonic Acids , Breast Feeding , Caprylates , Environmental Pollutants , Fluorocarbons , Maternal Exposure , Humans , Fluorocarbons/blood , Alkanesulfonic Acids/blood , Female , Caprylates/blood , Pregnancy , Child , Child, Preschool , Infant , Environmental Pollutants/blood , Maternal Exposure/statistics & numerical data , Infant, Newborn , Male , Environmental Exposure/analysis , Diet , Prenatal Exposure Delayed Effects , Adult
6.
Eur Thyroid J ; 13(3)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38657654

ABSTRACT

Objective: The aim was to evaluate the possible association between some endocrine disruptive chemicals and thyroid cancer (TC) in an Italian case-control cohort. Methods: We enrolled 112 TC patients and 112 sex- and age-matched controls without known thyroid diseases. Per- and poly-fluoroalkyl substances (PFAS), poly-chlorinated biphenyls (PCBs), and dichlorodiphenyltrichloroethane (4,4'-DDT and 4,4'-DDE) were measured in the serum by liquid or gas chromatography-mass spectrometry. Unconditional logistic regression, Bayesan kernel machine regression and weighted quantile sum models were used to estimate the association between TC and pollutants' levels, considered individually or as mixture. BRAFV600E mutation was assessed by standard methods. Results: The detection of perfluorodecanoic acid (PFDA) was positively correlated to TC (OR = 2.03, 95% CI: 1.10-3.75, P = 0.02), while a negative association was found with perfluorohexanesulfonic acid (PFHxS) levels (OR = 0.63, 95% CI: 0.41-0.98, P = 0.04). Moreover, perfluorononanoic acid (PFNA) was positively associated with the presence of thyroiditis, while PFHxS and perfluorooctane sulfonic acid (PFOS) with higher levels of presurgical thyroid-stimulating hormone (TSH). PFHxS, PFOS, PFNA, and PFDA were correlated with less aggressive TC, while poly-chlorinated biphenyls (PCB-105 and PCB-118) with larger and more aggressive tumors. Statistical models showed a negative association between pollutants' mixture and TC. BRAF V600E mutations were associated with PCB-153, PCB-138, and PCB-180. Conclusion: Our study suggests, for the first time in a case-control population, that exposure to some PFAS and PCBs associates with TC and some clinical and molecular features. On the contrary, an inverse correlation was found with both PFHxS and pollutants' mixture, likely due to a potential reverse causality.


Subject(s)
Alkanesulfonic Acids , Endocrine Disruptors , Fluorocarbons , Persistent Organic Pollutants , Polychlorinated Biphenyls , Thyroid Neoplasms , Humans , Case-Control Studies , Fluorocarbons/blood , Fluorocarbons/adverse effects , Female , Male , Middle Aged , Endocrine Disruptors/blood , Endocrine Disruptors/adverse effects , Thyroid Neoplasms/epidemiology , Thyroid Neoplasms/blood , Thyroid Neoplasms/chemically induced , Thyroid Neoplasms/genetics , Polychlorinated Biphenyls/blood , Polychlorinated Biphenyls/adverse effects , Alkanesulfonic Acids/blood , Adult , Persistent Organic Pollutants/adverse effects , Persistent Organic Pollutants/blood , Aged , Dichlorodiphenyl Dichloroethylene/blood , Decanoic Acids/blood , Decanoic Acids/adverse effects , DDT/blood , DDT/adverse effects , Italy/epidemiology , Caprylates/blood , Caprylates/adverse effects , Proto-Oncogene Proteins B-raf/genetics , Fatty Acids/blood , Sulfonic Acids/blood , Mutation , Environmental Exposure/adverse effects
7.
Front Public Health ; 12: 1351786, 2024.
Article in English | MEDLINE | ID: mdl-38665245

ABSTRACT

Recent evidence has revealed associations between endocrine-disrupting chemicals (EDCs) and placental insufficiency due to altered placental growth, syncytialization, and trophoblast invasion. However, no epidemiologic study has reported associations between exposure to EDCs and asymmetric fetal growth restriction (FGR) caused by placenta insufficiency. The aim of this study was to evaluate the association between EDC exposure and asymmetric FGR. This was a prospective cohort study including women admitted for delivery to the Maternal Fetal Center at Seoul St. Mary's Hospital between October 2021 and October 2022. Maternal urine and cord blood samples were collected, and the levels of bisphenol-A (BPA), monoethyl phthalates, and perfluorooctanoic acid in each specimen were analyzed. We investigated linear and non-linear associations between the levels of EDCs and fetal growth parameters, including the head circumference (HC)/abdominal circumference (AC) ratio as an asymmetric parameter. The levels of EDCs were compared between fetuses with and without asymmetric FGR. Of the EDCs, only the fetal levels of BPA showed a linear association with the HC/AC ratio after adjusting for confounding variables (ß = 0.003, p < 0.05). When comparing the normal growth and asymmetric FGR groups, the asymmetric FGR group showed significantly higher maternal and fetal BPA levels compared to the normal growth group (maternal urine BPA, 3.99 µg/g creatinine vs. 1.71 µg/g creatinine [p < 0.05]; cord blood BPA, 1.96 µg/L vs. -0.86 µg/L [p < 0.05]). In conclusion, fetal exposure levels of BPA show linear associations with asymmetric fetal growth patterns. High maternal and fetal exposure to BPA might be associated with asymmetric FGR.


Subject(s)
Benzhydryl Compounds , Endocrine Disruptors , Fetal Blood , Fetal Growth Retardation , Maternal Exposure , Phenols , Humans , Female , Endocrine Disruptors/adverse effects , Endocrine Disruptors/blood , Endocrine Disruptors/urine , Prospective Studies , Pregnancy , Fetal Growth Retardation/chemically induced , Adult , Benzhydryl Compounds/adverse effects , Benzhydryl Compounds/urine , Benzhydryl Compounds/blood , Phenols/urine , Phenols/adverse effects , Phenols/blood , Maternal Exposure/adverse effects , Fetal Blood/chemistry , Fluorocarbons/blood , Fluorocarbons/adverse effects , Phthalic Acids/urine , Phthalic Acids/adverse effects , Caprylates/blood , Caprylates/adverse effects , Placental Insufficiency , Republic of Korea/epidemiology , Seoul/epidemiology
8.
Int J Hyg Environ Health ; 259: 114380, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38657330

ABSTRACT

BACKGROUND/AIMS: Pregnant women are exposed to persistent environmental contaminants, including per- and polyfluoroalkyl substances (PFAS) that disrupt thyroid function. However, it is unclear if PFAS alter maternal sex-steroid hormone levels, which support pregnancy health and fetal development. METHODS: In Illinois women with relatively high socioeconomic status (n = 460), we quantified perfluorononanoic (PFNA), perfluorooctane sulfonic (PFOS), perfluorooctanoic (PFOA), methyl-perfluorooctane sulfonamide acetic acid, perfluorohexanesulphonic (PFHxS), perfluorodecanoic (PFDeA), and perfluoroundecanoic (PFUdA) acid concentrations in fasting serum samples at median 17 weeks gestation, along with plasma progesterone, testosterone, and estradiol. We evaluated covariate-adjusted associations of ln-transformed hormones with each ln-transformed PFAS individually using linear regression and with the PFAS mixture using quantile-based g-computation (QGComp). RESULTS: Interquartile range (IQR) increases in PFOS were associated with higher progesterone (%Δ 3.0; 95%CI: -0.6, 6.6) and estradiol (%Δ: 8.1; 95%CI: 2.2, 14.4) levels. Additionally, PFHxS was positively associated with testosterone (%Δ: 10.2; 95%CI: 4.0, 16.7), whereas both PFDeA and PFUdA were inversely associated with testosterone (%Δ: -5.7; 95%CI: -10.3, -0.8, and %Δ: -4.1; 95%CI: -7.6, -0.4, respectively). The IQR-standardized PFAS mixture was not associated with progesterone (%Δ: 1.6; 95%CI: -5.8, 9.2), due equal partial positive (%Δ: 9.2; driven by PFOA) and negative (%Δ: -7.4; driven by PFOS) mixture associations. Similarly, the mixture was not associated with testosterone (%Δ: 5.3; 95%CI: -9.0, 20.1), due to similar partial positive (%Δ: 23.6; driven by PFHxS) and negative (%Δ: -17.4; driven by PFDeA) mixture associations. However, we observed a slightly stronger partial positive (%Δ: 25.6; driven by PFOS and PFUdA) than negative (%Δ: -16.3; driven by PFOA) association resulting in an overall non-significant positive trend between the mixture and estradiol (%Δ: 8.5; 95%CI: -3.7, 20.9). CONCLUSION: PFAS mixture modeled using QGComp was not associated with maternal sex-steroid hormones due to potential opposing effects of certain PFAS. Additional prospective studies could corroborate these findings.


Subject(s)
Alkanesulfonic Acids , Environmental Pollutants , Fluorocarbons , Pregnancy Trimester, Second , Female , Humans , Fluorocarbons/blood , Pregnancy , Adult , Environmental Pollutants/blood , Pregnancy Trimester, Second/blood , Alkanesulfonic Acids/blood , Estradiol/blood , Young Adult , Illinois , Gonadal Steroid Hormones/blood , Testosterone/blood , Progesterone/blood , Fatty Acids/blood , Caprylates/blood , Maternal Exposure
9.
Int J Hyg Environ Health ; 259: 114385, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38676994

ABSTRACT

AIMS: Recent epidemiologic research has examined the relationship between perfluoroalkyl and polyfluoroalkyl substances (PFAS) and diabetes mellitus with inconclusive findings. In this cross-sectional study, we aimed to explore the association between serum PFAS concentrations and the prevalence of prediabetes and pre-diagnostic diabetes in the general Korean population as well as the combined effects of exposure to mixed PFAS compounds. METHODS: We analyzed data from participants aged ≥19 years enrolled in the Korean National Environmental Health Survey Cycle 4 (2018-2020). Individuals diagnosed with diabetes were excluded to minimize potential bias. We identified cases of pre-diagnostic diabetes based on the HbA1c level ≥6.5% and prediabetes as HbA1c levels of 5.7-6.49%. Serum concentrations of PFAS, including perfluorooctanoic acid (PFOA), perfluorononanoic acid (PFNA), perfluorodecanoic acid (PFDeA), perfluorohexane sulfonic acid (PFHxS), and perfluorooctane sulfonic acid (PFOS), were quantified using high-performance liquid chromatography-tandem mass spectrometry. Survey-weighted logistic regression models were used to assess the relationships between PFAS levels and diabetes risk, adjusting for covariates. Additionally, Bayesian kernel machine regression (BKMR) was used to investigate the combined effects of exposure to mixed PFAS compounds. RESULTS: In the study population excluding participants with diagnosed diabetes (n = 2709), the prevalence of pre-diagnostic diabetes and prediabetes was 4.8% and 30.1%, respectively. Significant positive associations were found between serum PFHxS and PFOS quartiles and pre-diagnostic diabetes risk. Likewise, among those without diagnosed or pre-diagnostic diabetes (n = 2579), the highest quartiles of PFDeA, PFHxS, and PFOS and the overall PFAS level were associated with an increased risk of prediabetes compared with the lowest quartiles. BKMR analysis revealed a significant positive association between overall serum PFAS level and prediabetes risk, which was most marked for PFOS. CONCLUSIONS: These findings highlight the potential health implications of PFAS exposure and prediabetes risk. Further research is needed to validate these associations and identify potential mechanistic pathways.


Subject(s)
Alkanesulfonic Acids , Diabetes Mellitus , Environmental Pollutants , Fluorocarbons , Humans , Fluorocarbons/blood , Middle Aged , Female , Republic of Korea/epidemiology , Male , Diabetes Mellitus/epidemiology , Diabetes Mellitus/blood , Adult , Environmental Pollutants/blood , Alkanesulfonic Acids/blood , Cross-Sectional Studies , Aged , Health Surveys , Environmental Exposure/adverse effects , Prevalence , Caprylates/blood , Prediabetic State/blood , Prediabetic State/epidemiology , Decanoic Acids/blood , Young Adult , Fatty Acids
10.
Chemosphere ; 357: 142052, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38631500

ABSTRACT

BACKGROUND: Per- and polyfluoroalkyl substances (PFAS) are man-made chemicals that are slow to break down in the environment and widely detected in humans. Epidemiological evidence suggests that prenatal exposure to perfluorooctanoic acid (PFOA), a legacy PFAS, is linked to gestational hypertension and preeclampsia. However, the relationship between other PFAS, which are structurally similar, and these outcomes remains largely understudied, despite biologic plausibility. Here, we examined associations between serum PFAS mixtures in relation to hypertensive disorders of pregnancy within a birth cohort of African Americans. METHODS: Participants in the present study were enrolled in the Atlanta African American Maternal-Child cohort between 2014 and 2020 (n = 513). Serum samples collected between 8 and 14 weeks gestation were analyzed for four PFAS. Logistic regression was used to assess associations between individual natural log transformed PFAS and specific hypertensive disorders of pregnancy (preeclampsia, gestational hypertension), while quantile g-computation was used to estimate mixture effects. Preeclampsia and gestational hypertension were treated as separate outcomes in individual models. All models were adjusted for maternal education, maternal age, early pregnancy body mass index, parity, and any alcohol, tobacco, or marijuana use. RESULTS: The geometric mean of PFOS and PFHxS was slightly lower among those with preeclampsia relative to those without a hypertensive disorder (e.g., geometric mean for PFOS was 1.89 and 1.94, respectively). Serum concentrations of PFAS were not strongly associated with gestational hypertension or preeclampsia in single pollutant or mixture models. For example, using quantile g-computation, a simultaneous one quartile increase in all PFAS was not associated with odds of gestational hypertension (odds ratio = 0.86, 95% CI = 0.60, 1.23), relative to those without a hypertensive disorder of pregnancy. CONCLUSIONS: In this birth cohort of African Americans, there was no association between serum PFAS measured in early pregnancy and hypertensive disorders of pregnancy, which may be reflective of the fairly low PFAS levels in our study population.


Subject(s)
Black or African American , Environmental Pollutants , Fluorocarbons , Hypertension, Pregnancy-Induced , Maternal Exposure , Humans , Female , Fluorocarbons/blood , Pregnancy , Black or African American/statistics & numerical data , Adult , Hypertension, Pregnancy-Induced/epidemiology , Hypertension, Pregnancy-Induced/blood , Maternal Exposure/statistics & numerical data , Environmental Pollutants/blood , Cohort Studies , Caprylates/blood , Georgia/epidemiology , Young Adult , Prenatal Exposure Delayed Effects , Pre-Eclampsia/blood , Pre-Eclampsia/epidemiology , Alkanesulfonic Acids/blood
11.
Sci Total Environ ; 928: 172316, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38593875

ABSTRACT

Per- and polyfluoroalkyl substances (PFAS) are fluorinated organic compounds used in a variety of consumer products and industrial applications that persist in the environment, bioaccumulate in biological tissues, and can have adverse effects on human health, especially in vulnerable populations. In this study, we focused on PFAS exposures in residents of senior care facilities. To investigate relationships between indoor, personal, and internal PFAS exposures, we analyzed 19 PFAS in matched samples of dust collected from the residents' bedrooms, and wristbands and serum collected from the residents. The median ∑PFAS concentrations (the sum of all PFAS detected in the samples) measured in dust, wristbands, and serum were 120 ng/g, 0.05 ng/g, and 4.0 ng/mL, respectively. The most abundant compounds in serum were linear- and branched-perfluorooctane sulfonic acid (L-PFOS and B-PFOS, respectively) at medians of 1.7 ng/mL and 0.83 ng/mL, respectively, followed by the linear perfluorooctanoic acid (L-PFOA) found at a median concentration of 0.59 ng/mL. Overall, these three PFAS comprised 80 % of the serum ∑PFAS concentrations. A similar pattern was observed in dust with L-PFOS and L-PFOA found as the most abundant PFAS (median concentrations of 13 and 7.8 ng/g, respectively), with the overall contribution of 50 % to the ∑PFAS concentration. Only L-PFOA was found in wristbands at a median concentration of 0.02 ng/g. Significant correlations were found between the concentrations of several PFAS in dust and serum, and in dust and wristbands, suggesting that the indoor environment could be a significant contributor to the personal and internal PFAS exposures in seniors. Our findings demonstrate that residents of assisted living facilities are widely exposed to PFAS, with several PFAS found in blood of each study participant and in the assisted living environment.


Subject(s)
Environmental Exposure , Fluorocarbons , Fluorocarbons/analysis , Fluorocarbons/blood , Humans , Aged , Environmental Exposure/statistics & numerical data , Environmental Exposure/analysis , Dust/analysis , Environmental Pollutants/blood , Environmental Pollutants/analysis , Environmental Monitoring , Female , Male , Alkanesulfonic Acids/blood , Alkanesulfonic Acids/analysis , Aged, 80 and over , Caprylates/blood , Caprylates/analysis , Homes for the Aged/statistics & numerical data
12.
Am J Epidemiol ; 193(6): 917-925, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38400650

ABSTRACT

Few methods have been used to characterize repeatedly measured biomarkers of chemical mixtures. We applied latent profile analysis (LPA) to serum concentrations of 4 perfluoroalkyl and polyfluoroalkyl substances (PFAS) measured at 4 time points from gestation to age 12 years. We evaluated the relationships between profiles and z scores of height, body mass index, fat mass index, and lean body mass index at age 12 years (n = 218). We compared LPA findings with an alternative approach for cumulative PFAS mixtures using g-computation to estimate the effect of simultaneously increasing the area under the receiver operating characteristic curve (AUC) for all PFAS. We identified 2 profiles: a higher PFAS profile (35% of sample) and a lower PFAS profile (relative to each other), based on their average PFAS concentrations at all time points. The higher PFAS profile had generally lower z scores for all outcomes, with somewhat larger effects for males, though all 95% CIs crossed the null. For example, the higher PFAS profile was associated with a 0.50-unit lower (ß = -0.50; 95% CI, -1.07 to 0.08) BMI z score among males but not among females (ß = 0.04; 95% CI, -0.45 to 0.54). We observed similar patterns with AUCs. We found that a higher childhood PFAS profile and higher cumulative PFAS mixtures may be associated with altered growth in early adolescence. This article is part of a Special Collection on Environmental Epidemiology.


Subject(s)
Body Composition , Body Mass Index , Environmental Exposure , Fluorocarbons , Humans , Fluorocarbons/blood , Female , Male , Child , Body Composition/drug effects , Environmental Exposure/adverse effects , Environmental Exposure/analysis , Longitudinal Studies , Pregnancy , Adolescent , Environmental Pollutants/blood , Alkanesulfonic Acids/blood , Caprylates/blood , Prenatal Exposure Delayed Effects , Child, Preschool
14.
Article in English | MEDLINE | ID: mdl-35114474

ABSTRACT

Perfluorooctanoic acid (PFOA) and perfluorooctanesulfonic acid (PFOS) are synthetic chemicals that have been used in various industries and household products. These can easily accumulate in the human body, causing adverse effects on human health. In this study, a high-performance liquid chromatography-tandem mass spectrometry method was developed and validated for the simultaneous analysis of PFOA and linear PFOS in human serum. Owing to a lack of PFOA- and PFOS-free human serum, 13C8-PFOA and 13C8-PFOS were used as surrogate analytes for quantification. A sensitive and selective sample preparation method was developed and optimized by combining solid-phase extraction and protein precipitation method. The lower limit of quantification was 0.05 ng/mL, and the analytical response was linear up to 10 ng/mL for both PFOA and linear PFOS. Chromatographic separation of the linear PFOS from branched isomers was achieved within 5.5 min. The method was validated at various concentrations and afforded acceptable accuracy and precision values. After validation, the method was successfully applied to evaluate the exposure levels of PFOA and linear PFOS in the Korean population. The serum concentrations of PFOA and linear PFOS were 0.42-28.3 ng/mL and 0.81-57.6 ng/mL, respectively. The median concentration of linear PFOS was approximately 2.6-fold higher than that of PFOA. The concentration of PFOA was higher in women than men (p < 0.05) and that of linear PFOS was not significantly different between men and women. Therefore, a sensitive, selective, and reliable bioanalytical method was developed and validated. This method can potentially be applied to biomonitoring studies involving PFOA and linear PFOS.


Subject(s)
Alkanesulfonic Acids/blood , Caprylates/blood , Chromatography, High Pressure Liquid/methods , Fluorocarbons/blood , Tandem Mass Spectrometry/methods , Adolescent , Adult , Aged , Environmental Exposure/analysis , Female , Humans , Limit of Detection , Linear Models , Male , Middle Aged , Reproducibility of Results , Republic of Korea , Young Adult
15.
Reprod Toxicol ; 107: 22-32, 2022 01.
Article in English | MEDLINE | ID: mdl-34801790

ABSTRACT

We assessed how the interaction between mono-(2-ethylhexyl) phthalate (MEHP) in maternal sera and the maternal genotypes associated with nuclear receptors affect fatty acid levels in a prospective birth cohort study of pregnant Japanese individuals (n = 437) recruited in Sapporo between 2002 and 2005. We analyzed MEHP and fatty acids using gas chromatography-mass spectrometry. Thirteen single nucleotide polymorphisms of peroxisome proliferator-activated receptor (PPAR) alpha, PPAR gamma (PPARG), PPARG coactivator 1A (PPARGC1A), PPAR delta, constitutive androstane receptor, liver X receptor (LXR) alpha, and LXR beta (LXRB) were analyzed using real-time PCR. Multiple linear regression models were used to confirm the influence of log10-transformed MEHP levels and maternal genotypes on log10-transformed fatty acid levels. When the effects of the interaction between MEHP levels and the maternal PPARGC1A (rs8192678) genotype on oleic acid levels were evaluated, the estimated changes (95 % confidence intervals) in oleic acid levels against MEHP levels, maternal PPARGC1A (rs8192678)-GA/AA genotype, and the interaction between them showed a mean reduction of 0.200 (0.079, 0.322), mean reduction of 0.141 (0.000, 0.283), and mean increase of 0.145 (0.010, 0.281), respectively, after adjusting for the perfluorooctanesulfonate level. The effects of the interaction between MEHP levels and maternal LXRB (rs2303044) genotype on linoleic acid levels was also significant (pint = 0.010). In conclusion, the interaction between MEHP and the maternal genotypes PPARGC1A (rs8192678) and LXRB (rs2303044) decreased fatty acid levels. Further, the interaction between MEHP and PPARGC1A (rs8192678) may have a greater effect on fatty acid levels than the interaction between PFOS and PPARGC1A.


Subject(s)
Diethylhexyl Phthalate/analogs & derivatives , Environmental Pollutants/blood , Fatty Acids/blood , Receptors, Cytoplasmic and Nuclear/genetics , Adult , Alkanesulfonic Acids/blood , Asian People/genetics , Caprylates/blood , Diethylhexyl Phthalate/blood , Female , Fluorocarbons/blood , Genotype , Humans , Japan , Pregnancy
16.
Reprod Toxicol ; 107: 112-122, 2022 01.
Article in English | MEDLINE | ID: mdl-34896592

ABSTRACT

We assessed the associations between perfluorooctanesulfonate (PFOS) and perfluorooctanoate (PFOA) levels in third trimester maternal serum, the maternal genotypes of genes encoding nuclear receptors, and birth outcomes. We studied a prospective birth cohort of healthy pregnant Japanese women (n = 372) recruited in Sapporo between July 2002 and October 2005. We analyzed PFOS and PFOA levels using liquid chromatography-tandem mass spectrometry and analyzed 13 single nucleotide polymorphisms (SNPs) of proliferator-activated receptor alpha, gamma, gamma coactivator 1A, delta, constitutive androstane receptor, liver X receptor alpha, and beta (LXRB) using real-time polymerase reaction (PCR). We employed multiple linear regression models to establish the influences of log10-transformed PFOS and PFOA levels and maternal genotypes on birth size. In female infants, we identified interactions between PFOS levels, the maternal genotype of LXRB (rs1405655), and birth weight. The estimated mean changes in birth weight in response to PFOS levels, the maternal genotype LXRB (rs1405655)-TC/CC (compared to TT), and their interactions were -502.9 g (95 % confidence interval [CI] = -247.3, -758.5 g), -526.3 g (95 % CI = -200.7, -852.0 g), and 662.1 g (95 % CI = 221.0, 1,103.2 g; pint = 0.003), respectively. Interactions between PFOS levels and the maternal genotype of LXRB (rs1405655) also significantly affected birth chest circumference and the Ponderal index (pint = 0.037 and 0.005, respectively). Thus, interactions between PFOS levels and the maternal genotype of LXRB (rs1405655) affects birth sizes in female infants. We found that certain SNPs modify the effects of PFOS levels on birth size.


Subject(s)
Alkanesulfonic Acids/blood , Birth Weight , Caprylates/blood , Environmental Pollutants/blood , Fluorocarbons/blood , Receptors, Cytoplasmic and Nuclear/genetics , Adult , Birth Cohort , Cohort Studies , Fatty Acids/blood , Female , Humans , Infant, Newborn , Japan/epidemiology , Male , Polymorphism, Single Nucleotide , Pregnancy , Pregnancy Complications/blood , Pregnancy Complications/genetics , Young Adult
17.
Sci Rep ; 11(1): 19789, 2021 10 05.
Article in English | MEDLINE | ID: mdl-34611219

ABSTRACT

Prenatal exposure to perfluoroalkyl substances (PFAS) has been reported to affect body weight from birth to childhood, but the results remain inconclusive. We investigated whether umbilical cord blood concentrations of perfluorooctane sulfonate (PFOS) and perfluorooctanoic acid (PFOA) are associated with children's risk trajectory for obesity. 600 children were randomly selected from the Hamamatsu Birth Cohort for Mothers and Children (HBC study) and their umbilical cord serum PFAS concentrations were quantified. Participants underwent BMI measurements at ages 1, 4, 10, 18, 24, 32, 40, 50, and 66 months. Growth curve modeling with random intercept was performed with standardized BMI as outcome variable. PFOS was negatively associated with standardized BMI (ß = - 0.34; p = 0.01), with a marginally significant interaction with the child's age (ß = 0.0038; p = 0.08). PFOA was negatively associated with standardized BMI (ß = - 0.26, 95% CI - 0.51, 0; p = 0.05), with a significant interaction with the child's age (ß = 0.005; p = 0.01). Stratified analysis by sex revealed that these effects were significant only among girls. Prenatal exposure to PFAS initially was associated with lower standardized BMI during infancy, but this effect dissipated over time and reversed in direction during later childhood. The effects of prenatal PFAS on higher standardized BMI is stronger in girls.


Subject(s)
Alkanesulfonic Acids/blood , Body Mass Index , Caprylates/blood , Fetal Blood , Fluorocarbons/blood , Maternal Exposure/adverse effects , Prenatal Exposure Delayed Effects , Birth Weight , Child Development , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male , Pregnancy
18.
Reprod Toxicol ; 105: 221-231, 2021 10.
Article in English | MEDLINE | ID: mdl-34536543

ABSTRACT

Prenatal sex hormones affect fetal growth; for example, prenatal exposure to low levels of androgen accelerates female puberty onset. We assessed the association of perfluoroalkyl substances (PFASs) in maternal sera and infant genotypes of genes encoding enzymes involved in sex steroid hormone biosynthesis on cord sera sex hormone levels in a prospective birth cohort study of healthy pregnant Japanese women (n = 224) recruited in Sapporo between July 2002 and October 2005. We analyzed PFAS and five sex hormone levels using liquid chromatography-tandem mass spectrometry. Cytochrome P450 (CYP) 17A1 (CYP17A1 rs743572), 19A1 (CYP19A1 rs10046, rs700519, and rs727479), 3ß-hydroxysteroid dehydrogenase type 1 (HSD3B1 rs6203), type 2 (HSD3B2 rs1819698, rs2854964, and rs4659175), 17ß-hydroxysteroid dehydrogenase type 1 (HSD17B1 rs605059, rs676387, and rs2676531), and type 3 (HSD17B3 rs4743709) were analyzed using real-time PCR. Multiple linear regression models were used to establish the influence of log10-transformed PFAS levels and infant genotypes on log10-transformed sex steroid hormone levels. When the interaction between perfluorooctanesulfonate (PFOS) levels and female infant genotype CYP17A1 (rs743572) on the androstenedione (A-dione) levels was considered, the estimated changes (95 % confidence intervals) in A-dione levels against PFOS levels, female infant genotype CYP17A1 (rs743572)-AG/GG, and interaction between them showed a mean increase of 0.445 (0.102, 0.787), mean increase of 0.392 (0.084, 0.707), and mean reduction of 0.579 (0.161, 0.997) (Pint = 0.007), respectively. Moreover, a female-specific interaction with testosterone levels was observed. A-dione and T levels showed positive main effects and negative interaction with PFOS levels and the female infant CYP17A1 genotype.


Subject(s)
Alkanesulfonic Acids/blood , Caprylates/blood , Cytochrome P-450 Enzyme System/genetics , Environmental Pollutants/blood , Fluorocarbons/blood , Gonadal Steroid Hormones/blood , Hydroxysteroid Dehydrogenases/genetics , Adult , Female , Fetal Blood/chemistry , Fetus , Genotype , Humans , Infant, Newborn , Male , Maternal Exposure , Maternal-Fetal Exchange , Polymorphism, Genetic , Pregnancy
19.
J Clin Endocrinol Metab ; 106(11): e4427-e4437, 2021 10 21.
Article in English | MEDLINE | ID: mdl-34181018

ABSTRACT

CONTEXT: Per- and polyfluoroalkyl substances (PFAS) are widespread chemicals that may affect sex hormones and accelerate reproductive aging in midlife women. OBJECTIVE: To examine associations between serum PFAS concentrations at baseline (1999-2000) and longitudinal serum concentrations of follicle-stimulating hormone (FSH), estradiol, testosterone, and sex hormone-binding globulin (SHBG) at baseline and through 2015-2016. DESIGN: Prospective cohort. SETTING: General community. PARTICIPANTS: 1371 midlife women 45 to 56 years of age at baseline in the Study of Women's Health Across the Nation (SWAN). MAIN OUTCOME MEASURE(S): FSH, estradiol, testosterone, SHBG. RESULTS: In linear mixed models fitted with log-transformed hormones and log-transformed PFAS adjusting for age, site, race/ethnicity, smoking status, menopausal status, parity, and body mass index, FSH was positively associated with linear perfluorooctanoate [n-PFOA; 3.12% (95% CI 0.37%, 5.95%) increase for a doubling in serum concentration), linear perfluorooctane sulfonate [PFOS; 2.88% (0.21%, 5.63%)], branched perfluorooctane sulfonate [2.25% (0.02%, 4.54%)], total PFOS (3.03% (0.37%, 5.76%)), and 2-(N-ethyl-perfluorooctane sulfonamido) acetate [EtFOSAA; 1.70% (0.01%, 3.42%)]. Estradiol was inversely associated with perfluorononanoate [PFNA; -2.47% (-4.82%, -0.05%)) and n-PFOA (-2.43% (-4.97%, 0.18%)]. Significant linear trends were observed in the associations between PFOS and EtFOSAA with SHBG across parity (Ps trend ≤ 0.01), with generally inverse associations among nulliparous women but positive associations among women with 3+ births. No significant associations were observed between PFAS and testosterone. CONCLUSIONS: This study observed positive associations of PFOA and PFOS with FSH and inverse associations of PFNA and PFOA with estradiol in midlife women during the menopausal transition, consistent with findings that PFAS affect reproductive aging.


Subject(s)
Alkanesulfonic Acids/blood , Caprylates/blood , Fatty Acids/blood , Fluorocarbons/blood , Gonadal Steroid Hormones/blood , Menopause/blood , Cohort Studies , Estradiol/blood , Female , Follicle Stimulating Hormone/blood , Humans , Longitudinal Studies , Menopause/physiology , Middle Aged , Prospective Studies , Sex Hormone-Binding Globulin/analysis , Testosterone/blood , Women's Health
20.
Environ Health ; 20(1): 73, 2021 06 28.
Article in English | MEDLINE | ID: mdl-34187491

ABSTRACT

BACKGROUND: Exposure to perfluoroalkyl substances (PFASs) has been associated with changes in body mass index and adiposity, but evidence is inconsistent as study design, population age, follow-up periods and exposure levels vary between studies. We investigated associations between PFAS exposure and body fat in a cross-sectional study of healthy boys. METHODS: In 109 boys (10-14 years old), magnetic resonance imaging and dual-energy X-ray absorptiometry were performed to evaluate abdominal, visceral fat, total body, android, gynoid, android/gynoid ratio, and total fat percentage standard deviation score. Serum was analysed for perfluorooctanoic acid, perfluorooctane sulfonic acid (PFOS), perfluorohexane sulfonic acid, perfluorononanoic acid, and perfluorodecanoic acid using liquid chromatography and triple quadrupole mass spectrometry. Data were analysed by multivariate linear regression. RESULTS: Serum concentrations of PFASs were low. Generally, no clear associations between PFAS exposure and body fat measures were found; however, PFOS was negatively associated with abdominal fat (ß = -0.18, P = 0.046), android fat (ß = -0.34, P = 0.022), android/gynoid ratio (ß = -0.21, P = 0.004), as well as total body fat (ß = -0.21, P = 0.079) when adjusting for Tanner stage. CONCLUSIONS: Overall, we found no consistent associations between PFAS exposure and body fat. This could be due to our cross-sectional study design. Furthermore, we assessed PFAS exposure in adolescence and not in utero, which is considered a more vulnerable time window of exposure.


Subject(s)
Adipose Tissue , Alkanesulfonic Acids/blood , Caprylates/blood , Decanoic Acids/blood , Environmental Pollutants/blood , Fluorocarbons/blood , Absorptiometry, Photon , Adolescent , Biological Monitoring , Child , Humans , Magnetic Resonance Imaging , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...