Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.034
Filter
1.
Lancet Planet Health ; 8(5): e297-e308, 2024 May.
Article in English | MEDLINE | ID: mdl-38723642

ABSTRACT

BACKGROUND: Pregnancy air pollution exposure (PAPE) has been linked to a wide range of adverse birth and childhood outcomes, but there is a paucity of data on its influence on the placental epigenome, which can regulate the programming of physiological functions and affect child development. This study aimed to investigate the association between prenatal air pollutant exposure concentrations and changes in placental DNA methylation patterns, and to explore the potential windows of susceptibility and sex-specific alterations. METHODS: This multi-site study used three prospective population-based mother-child cohorts: EDEN, PELAGIE, and SEPAGES, originating from four French geographical regions (Nancy, Poitiers, Brittany, and Grenoble). Pregnant women were included between 2003 and 2006 for EDEN and PELAGIE, and between 2014 and 2017 for SEPAGES. The main eligibility criteria were: being older than 18 years, having a singleton pregnancy, and living and planning to deliver in one of the maternity clinics in one of the study areas. A total of 1539 mother-child pairs were analysed, measuring placental DNA methylation using Illumina BeadChips. We used validated spatiotemporally resolved models to estimate PM2·5, PM10, and NO2 exposure over each trimester of pregnancy at the maternal residential address. We conducted a pooled adjusted epigenome-wide association study to identify differentially methylated 5'-C-phosphate-G-3' (CpG) sites and regions (assessed using the Infinium HumanMethylationEPIC BeadChip array, n=871), including sex-specific and sex-linked alterations, and independently validated our results (assessed using the Infinium HumanMethylation450 BeadChip array, n=668). FINDINGS: We identified four CpGs and 28 regions associated with PAPE in the total population, 469 CpGs and 87 regions in male infants, and 150 CpGs and 66 regions in female infants. We validated 35% of the CpGs available. More than 30% of the identified CpGs were related to one (or more) birth outcome and most significant alterations were enriched for neural development, immunity, and metabolism related genes. The 28 regions identified for both sexes overlapped with imprinted genes (four genes), and were associated with neurodevelopment (nine genes), immune system (seven genes), and metabolism (five genes). Most associations were observed for the third trimester for female infants (134 of 150 CpGs), and throughout pregnancy (281 of 469 CpGs) and the first trimester (237 of 469 CpGs) for male infants. INTERPRETATION: These findings highlight the molecular pathways through which PAPE might affect child health in a widespread and sex-specific manner, identifying the genes involved in the major physiological functions of a developing child. Further studies are needed to elucidate whether these epigenetic changes persist and affect health later in life. FUNDING: French Agency for National Research, Fondation pour la Recherche Médicale, Fondation de France, and the Plan Cancer.


Subject(s)
Air Pollutants , Air Pollution , DNA Methylation , Maternal Exposure , Placenta , Humans , Female , Pregnancy , Placenta/drug effects , Placenta/metabolism , Prospective Studies , Maternal Exposure/adverse effects , Adult , Air Pollution/adverse effects , Male , Air Pollutants/adverse effects , Air Pollutants/analysis , France , Prenatal Exposure Delayed Effects/genetics , Pregnancy Outcome , Infant, Newborn , Young Adult
2.
Environ Health Perspect ; 132(5): 57002, 2024 May.
Article in English | MEDLINE | ID: mdl-38728218

ABSTRACT

BACKGROUND: Endocrine-disrupting chemicals may play a role in adiposity development during childhood. Until now literature in this scope suffers from methodologic limitations in exposure assessment using one or few urine samples and missing assessment during the infancy period. OBJECTIVES: We investigated the associations between early-life exposure to quickly metabolized chemicals and post-natal growth, relying on repeated within-subject urine collections over pregnancy and infancy. METHODS: We studied the associations of four phenols, four parabens, seven phthalates, and one nonphthalate plasticizer from weekly pooled urine samples collected from the mother during second and third trimesters (median 18 and 34 gestational weeks, respectively) and infant at 2 and 12 months of age, and child growth until 36 months. We relied on repeated measures of height, weight and head circumference from study visits and the child health booklet to predict growth outcomes at 3 and 36 months using the Jenss-Bayley nonlinear mixed model. We assessed associations with individual chemicals using adjusted linear regression and mixtures of chemicals using a Bayesian kernel machine regression model. RESULTS: The unipollutant analysis revealed few associations. Bisphenol S (BPS) at second trimester was positively associated with all infant growth parameters at 3 and 36 months, with similar patterns between exposure at third trimester and all infant growth parameters at 3 months. Mono-n-butyl phthalate (MnBP) at 12 months was positively associated with body mass index (BMI), weight, and head circumference at 36 months. Mixture analysis revealed positive associations between exposure at 12 months and BMI and weight at 36 months, with MnBP showing the highest effect size within the mixture. CONCLUSIONS: This study suggests that exposure in early infancy may be associated with increased weight and BMI in early childhood, which are risk factors of obesity in later life. Furthermore, this study highlighted the impact of BPS, a compound replacing bisphenol A, which has never been studied in this context. https://doi.org/10.1289/EHP13644.


Subject(s)
Endocrine Disruptors , Parabens , Phenols , Phthalic Acids , Prenatal Exposure Delayed Effects , Humans , Phthalic Acids/urine , Phenols/urine , Phenols/toxicity , Female , Infant , Pregnancy , Endocrine Disruptors/urine , Endocrine Disruptors/toxicity , Environmental Pollutants/urine , Male , Maternal Exposure/statistics & numerical data , Maternal Exposure/adverse effects , Longitudinal Studies , Child, Preschool , Anthropometry
3.
Int J Mol Sci ; 25(9)2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38731818

ABSTRACT

Early life exposure lays the groundwork for the risk of developing cardiovascular-kidney-metabolic (CKM) syndrome in adulthood. Various environmental chemicals to which pregnant mothers are commonly exposed can disrupt fetal programming, leading to a wide range of CKM phenotypes. The aryl hydrocarbon receptor (AHR) has a key role as a ligand-activated transcription factor in sensing these environmental chemicals. Activating AHR through exposure to environmental chemicals has been documented for its adverse impacts on cardiovascular diseases, hypertension, diabetes, obesity, kidney disease, and non-alcoholic fatty liver disease, as evidenced by both epidemiological and animal studies. In this review, we compile current human evidence and findings from animal models that support the connection between antenatal chemical exposures and CKM programming, focusing particularly on AHR signaling. Additionally, we explore potential AHR modulators aimed at preventing CKM syndrome. As the pioneering review to present evidence advocating for the avoidance of toxic chemical exposure during pregnancy and deepening our understanding of AHR signaling, this has the potential to mitigate the global burden of CKM syndrome in the future.


Subject(s)
Cardiovascular Diseases , Prenatal Exposure Delayed Effects , Receptors, Aryl Hydrocarbon , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Aryl Hydrocarbon/genetics , Humans , Pregnancy , Animals , Female , Prenatal Exposure Delayed Effects/metabolism , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/etiology , Cardiovascular Diseases/chemically induced , Kidney Diseases/chemically induced , Kidney Diseases/metabolism , Kidney Diseases/etiology , Maternal Exposure/adverse effects , Signal Transduction/drug effects , Kidney/metabolism , Kidney/drug effects , Kidney/pathology , Fetal Development/drug effects , Environmental Pollutants/toxicity , Environmental Pollutants/adverse effects , Metabolic Reprogramming
4.
BMJ Open ; 14(5): e079782, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38719310

ABSTRACT

PURPOSE: Pregnancy and the postpartum period are increasingly recognised as sensitive windows for cardiometabolic disease risk. Growing evidence suggests environmental exposures, including endocrine-disrupting chemicals (EDCs), are associated with an increased risk of pregnancy complications that are associated with long-term cardiometabolic risk. However, the impact of perinatal EDC exposure on subsequent cardiometabolic risk post-pregnancy is less understood. The Environmental Reproductive and Glucose Outcomes (ERGO) Study was established to investigate the associations of environmental exposures during the perinatal period with post-pregnancy parental cardiometabolic health. PARTICIPANTS: Pregnant individuals aged ≥18 years without pre-existing diabetes were recruited at <15 weeks of gestation from Boston, Massachusetts area hospitals. Participants completed ≤4 prenatal study visits (median: 12, 19, 26, 36 weeks of gestation) and 1 postpartum visit (median: 9 weeks), during which we collected biospecimens, health histories, demographic and behavioural data, and vitals and anthropometric measurements. Participants completed a postpartum fasting 2-hour 75 g oral glucose tolerance test. Clinical data were abstracted from electronic medical records. Ongoing (as of 2024) extended post-pregnancy follow-up visits occur annually following similar data collection protocols. FINDINGS TO DATE: We enrolled 653 unique pregnancies and retained 633 through delivery. Participants had a mean age of 33 years, 10% (n=61) developed gestational diabetes and 8% (n=50) developed pre-eclampsia. Participant pregnancy and postpartum urinary phthalate metabolite concentrations and postpartum glycaemic biomarkers were quantified. To date, studies within ERGO found higher exposure to phthalates and phthalate mixtures, and separately, higher exposure to radioactive ambient particulate matter, were associated with adverse gestational glycaemic outcomes. Additionally, certain personal care products used in pregnancy, notably hair oils, were associated with higher urinary phthalate metabolite concentrations, earlier gestational age at delivery and lower birth weight. FUTURE PLANS: Future work will leverage the longitudinal data collected on pregnancy and cardiometabolic outcomes, environmental exposures, questionnaires, banked biospecimens and paediatric data within the ERGO Study.


Subject(s)
Environmental Exposure , Humans , Female , Pregnancy , Adult , Prospective Studies , Boston/epidemiology , Environmental Exposure/adverse effects , Endocrine Disruptors/adverse effects , Endocrine Disruptors/urine , Young Adult , Glucose Tolerance Test , Blood Glucose/analysis , Blood Glucose/metabolism , Postpartum Period , Maternal Exposure/adverse effects , Cardiometabolic Risk Factors
5.
Rev Saude Publica ; 58: 19, 2024.
Article in English | MEDLINE | ID: mdl-38747867

ABSTRACT

OBJECTIVE: To identify the prevalence of contamination by pesticides and their metabolites in the milk of lactating mothers in Latin America. METHODS: In this systematic review, the PubMed, LILACS, Embase, and Scopus databases were searched up to January 2022 to identify observational studies. The Mendeley software was used to manage these references. The risk of bias assessment was evaluated according to the checklist for prevalence studies and writing design, by the Prisma guidelines. RESULTS: This study retrieved 1835 references and analyzed 49 studies. 69.38% of the analyzed studies found a 100% prevalence of breast milk contamination by pesticides among their sample. Main pesticides include dichlorodiphenyltrichloroethane (DDT) and its isomers (75.51%), followed by the metabolite dichlorodiphenyldichloroethylene (DDE) (69.38%) and hexachlorocyclohexane (HCH) (46.93%). This study categorized most (65.30%) studies as having a low risk of bias. CONCLUSIONS: This review shows a high prevalence of pesticide contamination in the breast milk of Latin American women. Further investigations should be carried out to assess contamination levels in breast milk and the possible effects of these substances on maternal and child health.


Subject(s)
Lactation , Milk, Human , Pesticides , Humans , Milk, Human/chemistry , Female , Latin America , Pesticides/analysis , Pesticide Residues/analysis , Prevalence , DDT/analysis , Maternal Exposure/adverse effects
6.
J Toxicol Sci ; 49(5): 209-218, 2024.
Article in English | MEDLINE | ID: mdl-38692908

ABSTRACT

The immune system is sensitive to many chemicals. Among dioxin compounds, 2,3,7,8-tetrachlorodizenzo-p-dioxin (TCDD) is the most toxic environmental pollutant. The effects of perinatal maternal exposure to dioxins may persist into childhood. However, there have been no reports to date on the effects of exposure to dioxins during infancy, when the immune organs are developing. Therefore, we investigated the effects of TCDD and antigen exposure during lactation on immune function, especially antibody production capacity, in adult mice. Beginning the day after delivery, lactating mothers were orally administered TCDD or a mixture of TCDD and ovalbumin (OVA) daily for 4 weeks, until the pups were weaned. At 6 weeks of age, progeny mice were orally administered OVA daily for 10 weeks, while non-progeny mice were orally administered OVA or a mixture of TCDD and OVA daily for 10 weeks. Production of serum OVA-specific IgG was examined weekly. The amount of TCDD transferred from the mother to the progeny via breast milk was determined by measuring TCDD in the gastric contents of the progeny. A trend toward increasing IgA titer was observed in TCDD-treated mice, and production of IgE was observed only in progeny whose mothers were treated with TCDD and OVA. The results suggest that exposure to TCDD and OVA in breast milk can affect immune function in newborns.


Subject(s)
Lactation , Ovalbumin , Polychlorinated Dibenzodioxins , Animals , Female , Ovalbumin/immunology , Ovalbumin/administration & dosage , Polychlorinated Dibenzodioxins/toxicity , Maternal Exposure/adverse effects , Antibody Formation/drug effects , Environmental Pollutants/toxicity , Immunoglobulin G/blood , Immunoglobulin A/blood , Immunoglobulin E/blood , Immunoglobulin E/immunology , Antigens/immunology , Mice , Pregnancy , Milk/immunology , Male , Milk, Human/immunology , Administration, Oral
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.
PLoS Med ; 21(4): e1004395, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38669277

ABSTRACT

BACKGROUND: Epidemiological findings regarding the association of particulate matter ≤2.5 µm (PM2.5) exposure with hypertensive disorders in pregnancy (HDP) are inconsistent; evidence for HDP risk related to PM2.5 components, mixture effects, and windows of susceptibility is limited. We aimed to investigate the relationships between HDP and exposure to PM2.5 during pregnancy. METHODS AND FINDINGS: A large retrospective cohort study was conducted among mothers with singleton pregnancies in Kaiser Permanente Southern California from 2008 to 2017. HDP were defined by International Classification of Diseases-9/10 (ICD-9/10) diagnostic codes and were classified into 2 subcategories based on the severity of HDP: gestational hypertension (GH) and preeclampsia and eclampsia (PE-E). Monthly averages of PM2.5 total mass and its constituents (i.e., sulfate, nitrate, ammonium, organic matter, and black carbon) were estimated using outputs from a fine-resolution geoscience-derived model. Multilevel Cox proportional hazard models were used to fit single-pollutant models; quantile g-computation approach was applied to estimate the joint effect of PM2.5 constituents. The distributed lag model was applied to estimate the association between monthly PM2.5 exposure and HDP risk. This study included 386,361 participants (30.3 ± 6.1 years) with 4.8% (17,977/373,905) GH and 5.0% (19,381/386,361) PE-E cases, respectively. In single-pollutant models, we observed increased relative risks for PE-E associated with exposures to PM2.5 total mass [adjusted hazard ratio (HR) per interquartile range: 1.07, 95% confidence interval (CI) [1.04, 1.10] p < 0.001], black carbon [HR = 1.12 (95% CI [1.08, 1.16] p < 0.001)] and organic matter [HR = 1.06 (95% CI [1.03, 1.09] p < 0.001)], but not for GH. The population attributable fraction for PE-E corresponding to the standards of the US Environmental Protection Agency (9 µg/m3) was 6.37%. In multi-pollutant models, the PM2.5 mixture was associated with an increased relative risk of PE-E ([HR = 1.05 (95% CI [1.03, 1.07] p < 0.001)], simultaneous increase in PM2.5 constituents of interest by a quartile) and PM2.5 black carbon gave the greatest contribution of the overall mixture effects (71%) among all individual constituents. The susceptible window is the late first trimester and second trimester. Furthermore, the risks of PE-E associated with PM2.5 exposure were significantly higher among Hispanic and African American mothers and mothers who live in low- to middle-income neighborhoods (p < 0.05 for Cochran's Q test). Study limitations include potential exposure misclassification solely based on residential outdoor air pollution, misclassification of disease status defined by ICD codes, the date of diagnosis not reflecting the actual time of onset, and lack of information on potential covariates and unmeasured factors for HDP. CONCLUSIONS: Our findings add to the literature on associations between air pollution exposure and HDP. To our knowledge, this is the first study reporting that specific air pollution components, mixture effects, and susceptible windows of PM2.5 may affect GH and PE-E differently.


Subject(s)
Air Pollution , Hypertension, Pregnancy-Induced , Particulate Matter , Humans , Female , Pregnancy , Retrospective Studies , Particulate Matter/adverse effects , Particulate Matter/analysis , Hypertension, Pregnancy-Induced/epidemiology , Hypertension, Pregnancy-Induced/etiology , Adult , Air Pollution/adverse effects , California/epidemiology , Air Pollutants/adverse effects , Air Pollutants/analysis , Young Adult , Maternal Exposure/adverse effects , Risk Factors , Environmental Exposure/adverse effects
9.
Environ Int ; 186: 108646, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38615543

ABSTRACT

Environmental exposures during pregnancy have been associated with adverse obstetric outcomes. However, limited and inconsistent evidence exists regarding the association between air temperature exposure and the risk of preeclampsia (PE). This study aimed to evaluate the correlation between ambient temperature exposure during pregnancy and PE risk, as well as identify the specific time window of temperature exposure that increases PE risk. A population-based cohort study was conducted from January 2012 to April 2022 in Guangzhou, China. Pregnant women were recruited in early pregnancy and followed until delivery. A total of 3,314 PE patients and 114,201 normal pregnancies were included. Ambient temperature exposures at different gestational weeks were recorded for each participant. Logistic regression models were used to evaluate the correlation between ambient temperature exposure and PE risk. Stratified analyses were conducted based on maternal age and pre-pregnancy BMI. Distributed lag models were employed to identify the time window of temperature exposure related to PE. Exposure to extreme high temperature (aOR = 1.24, 95 % CI 1.12-1.38) and moderate high temperature (aOR = 1.22, 95 % CI 1.10-1.35) during early pregnancy was associated with an increased risk of PE. Furthermore, women with higher pre-pregnancy BMI had a higher risk of developing PE when exposed to high temperature during early pregnancy compared to normal-weight women. The time window of temperature exposure related to PE was identified as pregnancy weeks 1 to 8. This study provides evidence for the association of high temperature exposure during early pregnancy with the risk of PE, as well as identifies the specific time window of temperature exposure related to PE. These findings have implications for developing potential strategies to protect pregnant women, particularly those with higher pre-pregnancy BMI, from the adverse effects of extreme temperatures during early pregnancy.


Subject(s)
Pre-Eclampsia , Temperature , Pregnancy , Humans , Female , Pre-Eclampsia/epidemiology , China/epidemiology , Adult , Environmental Exposure/statistics & numerical data , Cohort Studies , Risk Factors , Young Adult , Maternal Exposure/statistics & numerical data , Maternal Exposure/adverse effects
10.
Occup Environ Med ; 81(4): 209-216, 2024 Apr 28.
Article in English | MEDLINE | ID: mdl-38604660

ABSTRACT

BACKGROUND: There is inconsistent evidence of the effects of exposure to ambient air pollution on the occurrence of lower respiratory tract infections (LRTIs) in early childhood. We assessed the effects of individual-level prenatal and early life exposure to air pollutants on the risk of LRTIs in early life. METHODS: We studied 2568 members of the population-based Espoo Cohort Study born between 1984 and 1990 and living in 1991 in the City of Espoo, Finland. Exposure assessment was based on dispersion modelling and land-use regression for lifetime residential addresses. The outcome was a LRTI based on data from hospital registers. We applied Poisson regression to estimate the incidence rate ratio (IRR) of LTRIs, contrasting incidence rates in the exposure quartiles to the incidence rates in the first quartile. We used weighted quantile sum (WQS) regression to estimate the joint effect of the studied air pollutants. RESULTS: The risk of LRTIs during the first 2 years of life was significantly related to exposure to individual and multiple air pollutants, measured with the Multipollutant Index (MPI), including primarily sulphur dioxide (SO2), particulate matter with a dry diameter of up to 2.5 µm (PM2.5) and nitrogen dioxide (NO2) exposures in the first year of life, with an adjusted IRR of 1.72 per unit increase in MPI (95% CI 1.20 to 2.47). LRTIs were not related to prenatal exposure. CONCLUSIONS: We provide evidence that ambient air pollution exposure during the first year of life increases the risk of LRTIs during the first 2 years of life. SO2, PM2.5 and NO2 were found to contribute the highest weights on health effects.


Subject(s)
Air Pollutants , Air Pollution , Environmental Exposure , Nitrogen Dioxide , Particulate Matter , Prenatal Exposure Delayed Effects , Respiratory Tract Infections , Sulfur Dioxide , Humans , Pregnancy , Female , Prenatal Exposure Delayed Effects/epidemiology , Finland/epidemiology , Particulate Matter/adverse effects , Respiratory Tract Infections/epidemiology , Respiratory Tract Infections/etiology , Air Pollutants/adverse effects , Air Pollution/adverse effects , Air Pollution/analysis , Infant , Male , Nitrogen Dioxide/analysis , Nitrogen Dioxide/adverse effects , Child, Preschool , Cohort Studies , Environmental Exposure/adverse effects , Sulfur Dioxide/adverse effects , Sulfur Dioxide/analysis , Infant, Newborn , Incidence , Risk Factors , Adult , Maternal Exposure/adverse effects
11.
PLoS One ; 19(4): e0301144, 2024.
Article in English | MEDLINE | ID: mdl-38625962

ABSTRACT

INTRODUCTION: Noise exposure during pregnancy may affect a child's auditory system, which may disturb fetal learning and language development. We examined the impact of occupational noise exposure during pregnancy on children's language acquisition at the age of one. METHODS: A cohort study was conducted among women working in the food industry, as kindergarten teachers, musicians, dental nurses, or pharmacists who had a child aged <1 year. The analyses covered 408 mother-child pairs. Language acquisition was measured using the Infant-Toddler Checklist. An occupational hygienist assessed noise exposure individually as no (N = 180), low (70-78 dB; N = 108) or moderate/high exposure (>79 dB; N = 120). RESULTS: Among the boys, the adjusted mean differences in language acquisition scores were -0.4 (95% CI -2.5, 1.8) for low, and -0.7 (95% CI -2.9, 1.4) for moderate/high exposure compared to no exposure. Among the girls the respective scores were +0.1 (95% CI -2.2, 2.5) and -0.1 (95% CI -2.3, 2.2). Among the children of kindergarten teachers, who were mainly exposed to human noise, low or moderate exposure was associated with lower language acquisition scores. The adjusted mean differences were -3.8 (95% CI -7.2, -0.4) for low and -4.9 (95% CI -8.6, -1.2) for moderate exposure. CONCLUSIONS: In general, we did not detect an association between maternal noise exposure and children's language acquisition among one-year-old children. However, the children of kindergarten teachers exposed to human noise had lower language acquisition scores than the children of the non-exposed participants. These suggestive findings merit further investigation by level and type of exposure.


Subject(s)
Noise, Occupational , Occupational Exposure , Male , Pregnancy , Infant , Humans , Female , Cohort Studies , Noise, Occupational/adverse effects , Language Development , Maternal Exposure/adverse effects
12.
Toxicol Ind Health ; 40(6): 312-322, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38590048

ABSTRACT

Previous epidemiologic research has shown that phthalate exposure in pregnant women is related to adverse birth outcomes in a sex-specific manner. However, the biological mechanism of phthalate exposure that causes these birth outcomes remains poorly defined. In this research, we investigated the association between phthalate exposure and placental oxidative stress in a large population-based cohort study, aiming to initially explore the relationship between phthalate exposure and gene expression in placental oxidative stress in a sex-specific manner. Quantitative PCR was performed to measure the expression of placental inflammatory mRNAs (HO-1, HIF1α, and GRP78) in 2469 placentae. The multiple linear regression models were used to investigate the associations between mRNA and urinary phthalate monoesters. Phthalate metabolites monomethyl phthalate (MMP) and mono-n-butyl phthalate (MBP) were positively correlated with higher HIF1α expression in placentae of male fetuses (p < .05). Mono-benzyl phthalate (MBzP) increased the expression of HO-1, HIF1α, and GRP78 in placentae of male fetuses, and mono-(2-ethyl-5-hydroxyhexyl) phthalate (MEHHP) up-regulated the expression of HIF1α and GRP78. Additionally, mono-(2-ethyl-5-oxohexyl) phthalate (MEOHP) was negatively correlated with HO-1, HIF1α, and GRP78 in placentae of female fetuses. Maternal phthalate exposure was associated with oxidative stress variations in placental tissues. The associations were closer in the placentas of male fetuses than in that of female ones. The placenta oxidative stress is worth further investigation as a potential mediator of maternal exposure-induced disease risk in children.


Subject(s)
Biomarkers , Endoplasmic Reticulum Chaperone BiP , Maternal Exposure , Oxidative Stress , Phthalic Acids , Placenta , Humans , Phthalic Acids/toxicity , Phthalic Acids/urine , Female , Oxidative Stress/drug effects , Pregnancy , Male , Placenta/drug effects , Placenta/metabolism , Biomarkers/urine , Prospective Studies , Adult , Maternal Exposure/adverse effects , Sex Factors , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics , Cohort Studies
13.
J Hazard Mater ; 470: 134177, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38565010

ABSTRACT

Perfluorooctane sulfonates (PFOS) are the persistent organic pollutants. In the present study, 0, 0.3, or 3-mg/kg PFOS were administered to pregnant mice from GD 11 to GD 18. The histopathology of liver and intestine, serum and hepatic lipid levels, lipid metabolism related genes, and gut microbiota were examined in adult female offspring. The results suggested that maternal PFOS exposure increased serum levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), and induced F4/80+ macrophage infiltration in adult female offspring, in addition to the elevation of TNF-α and IL-1ß mRNA levels in low-dose and high-dose groups, respectively. Furthermore, maternal exposure to PFOS increased serum triglyceride (TG) and hepatic total cholesterol (TC) levels, which was associated with the alteration of the process of fatty acid transport and ß-oxidation, TG synthesis and transport, cholesterol synthesis and excretion in the liver. The AMPK/mTOR/autophagy signaling was also inhibited in the liver of adult female offspring. Moreover, changes in gut microbiota were also related to lipid metabolism, especially for the Desulfovibrio, Ligilactobacillus, Enterorhabdus, HT002 and Peptococcaceae_unclassified. Additionally, maternal exposure to PFOS decreased mRNA expressions of the tight junction protein and AB+ goblet cells in the colon, while increasing the overproduction of lipopolysaccharides (LPS) and F4/80+ macrophage infiltration. Collectively, maternal PFOS exposure induced liver lipid accumulation and inflammation, which strongly correlated with the disruption of the gut-liver axis and autophagy in adult female offspring, highlighting the persistent adverse effects in offspring exposed to PFOS.


Subject(s)
Alkanesulfonic Acids , Autophagy , Fluorocarbons , Gastrointestinal Microbiome , Lipid Metabolism , Liver , Maternal Exposure , Prenatal Exposure Delayed Effects , Animals , Fluorocarbons/toxicity , Female , Liver/drug effects , Liver/metabolism , Pregnancy , Gastrointestinal Microbiome/drug effects , Lipid Metabolism/drug effects , Alkanesulfonic Acids/toxicity , Autophagy/drug effects , Maternal Exposure/adverse effects , Inflammation/chemically induced , Mice , Male
14.
Sci Total Environ ; 928: 172411, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38608898

ABSTRACT

Exposure to diisodecyl phthalate (DIDP) during early pregnancy may be a risk factor for depressive behavior in offspring. While ozone (O3) exposure also raises the probability of depressive behavior during the preceding DIDP-induced process. In the present study, we investigated the effects of prenatal exposure to DIDP and O3 on the development of depressive-like behavior in offspring mice. The study found that prenatal exposure to both DIDP and O3 significantly increased depressive-like behavior in the offspring mice compared to either DIDP or O3 alone. Prenatal exposure to DIDP and O3 obviously increased the levels of corticotropin-releasing hormone (CRH), adrenocorticotropic hormone (ACTH) and cortisol, and decreased the levels of brain-derived neurotrophic factor (BDNF), 5-hydroxytryptamine (5-HT), dopamine (DA) and norepinephrine (NE) in the brain tissues of offspring mice. Transcriptome analysis further revealed significant alterations in genes related to oxidative stress and TWIST1 (a helix-loop-helix transcription factor) in response to the combined exposure to DIDP and O3. HPA axis activation, dysregulation of neurodevelopmental factors, oxidative stress and TWIST1 involvement, collectively contributed to the development of depression-like behaviors in offspring mice following prenatal exposure to DIDP and O3. Moreover, the study also verified the potential role of oxidative stress using vitamin E as an antioxidant. The findings provide valuable evidence for the relationship between co-exposure to DIDP and O3 and depression, highlighting the importance of considering the combined effects of multiple environmental pollutants in assessing their impact on mental health outcomes.


Subject(s)
Depression , Oxidative Stress , Ozone , Phthalic Acids , Prenatal Exposure Delayed Effects , Animals , Ozone/toxicity , Oxidative Stress/drug effects , Female , Pregnancy , Mice , Phthalic Acids/toxicity , Depression/chemically induced , Air Pollutants/toxicity , Behavior, Animal/drug effects , Nuclear Proteins/metabolism , Maternal Exposure/adverse effects
15.
Sci Total Environ ; 928: 172234, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38615756

ABSTRACT

BACKGROUND: Epidemiological studies have examined the relation between air pollution (NOx, NO2, PM2.5, PM2.5-10, and PM10) and adverse pregnancy outcomes (APOs). There's increasing evidence that air pollution increases the risk of APOs. However, the results of these studies are controversial, and the causal relation remains uncertain. We aimed to assess whether a genetic causal link exists between air pollution and APOs and the potential effects of this relation. METHODS: A novel two-sample Mendelian randomisation (MR) study used pooled data from a large-scale complete genome correlation study. The primary analysis method was inverse variance weighting (IVW), which explored the expose-outcome relationship for assessing single nucleotide polymorphisms (SNPs) associated with air pollution. Further sensitivity analysis, including MR-PRESSO, MR-Egger regression, and leave-one analysis, was used to test the consistency of the results. RESULTS: There was a significant correlation between air pollution-related SNPs and APOs. A robust causal link was found between genetic susceptibility to air pollution and APOs. CONCLUSIONS: Our MR analysis reveals a genetic causal relation between air pollution and APOs, which may help provide new insights into further mechanisms and clinical studies in air pollution-mediated APOs.


Subject(s)
Air Pollutants , Air Pollution , Mendelian Randomization Analysis , Polymorphism, Single Nucleotide , Pregnancy Outcome , Pregnancy , Female , Air Pollution/adverse effects , Air Pollution/statistics & numerical data , Humans , Pregnancy Outcome/epidemiology , Maternal Exposure/statistics & numerical data , Maternal Exposure/adverse effects , Particulate Matter , Environmental Exposure/statistics & numerical data , Environmental Exposure/adverse effects
16.
Ecotoxicol Environ Saf ; 276: 116311, 2024 May.
Article in English | MEDLINE | ID: mdl-38615639

ABSTRACT

Prenatal environmental exposure could be an essential health risk factor associated with neurodevelopmental disorders in offspring. However, the exact mechanisms underlying the impact of prenatal PM2.5 exposure on offspring cognition remain unclear. In our recent study using a PM2.5 exposed pregnant mouse model, we observed significant synaptic dysfunction in the hippocampi of the offspring. Concurrently, the epigenetic regulator of KDM5A and the Shh signaling pathway exhibited decreased activities. Significantly, changes in hippocampal KDM5A and Shh levels directly correlated with PM2.5 exposure intensity. Subsequent experiments revealed a marked reduction in the expression of Shh signaling and related synaptic proteins when KDM5A was silenced in cells. Notably, the effects of KDM5A deficiency were reversed significantly with the supplementation of a Shh activator. Furthermore, our findings indicate that Shh activation significantly attenuates PM2.5-induced synaptic impairments in hippocampal neurons. We further demonstrated that EGR1, a transcriptional inhibitor, plays a direct role in KDM5A's regulation of the Shh pathway under conditions of PM2.5 exposure. Our results suggest that the KDM5A's inhibitory regulation on the Shh pathway through the EGR1 gene is a crucial epigenetic mechanism underlying the synaptic dysfunction in hippocampal neurons caused by maternal PM2.5 exposure. This emphasizes the role of epigenetic regulations in neurodevelopmental disorders caused by environmental factors.


Subject(s)
Epigenesis, Genetic , Hedgehog Proteins , Hippocampus , Particulate Matter , Prenatal Exposure Delayed Effects , Signal Transduction , Hippocampus/drug effects , Animals , Female , Pregnancy , Signal Transduction/drug effects , Epigenesis, Genetic/drug effects , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Mice , Particulate Matter/toxicity , Retinoblastoma-Binding Protein 2/genetics , Maternal Exposure/adverse effects , Synapses/drug effects , Air Pollutants/toxicity
17.
Ecotoxicol Environ Saf ; 276: 116328, 2024 May.
Article in English | MEDLINE | ID: mdl-38636262

ABSTRACT

BACKGROUND: The relationships between maternal genetic and environmental exposure and conotruncal heart defects (CTDs) have been extensively investigated. Nevertheless, there is limited knowledge regarding the impact of ozone (O3) on the risk of CTDs. OBJECTIVE: To explore the correlation between maternal exposure to O3 and CTDs in China. METHODS: Pregnant women who underwent fetal echocardiography at Beijing Anzhen Hospital between January 2013 and December 2021 were enrolled. Their sociodemographic characteristics and lifestyle information, along with fetal data, were systematically collected. Fetal echocardiography was used to detect CTDs. Maternal exposure to ambient O3 during the embryonic period, the first trimester, the three months preceding the last menstrual period, and the perinatal period was estimated using residential addresses or hospital addresses associated with prenatal visits. The concentration of O3 was divided by quartiles, with the first quartile serving as a reference. Adjusted logistic regression models were employed to examine the associations between every 10 µg/m3 increase or quartile increase in ambient O3 exposure and CTDs. RESULTS: Among 24,278 subjects, 1069 exhibited fetuses with CTDs. Maternal exposure to ambient O3 during three pregnancy periods was associated with increased CTD risk. The adjusted odds ratio (OR) and 95% confidence interval (CI) were 1.271 (1.189-1.360) per 10 µg/m3 increase in O3 during the perinatal period. For each quartile of O3, the risk increased with increasing exposure concentration, particularly during the perinatal period (OR = 2.206 for quartile 2, 2.367 for quartile 3, and 3.378 for quartile 4, all P<0.05). CONCLUSIONS: Elevated maternal exposure to O3 during pregnancy, particularly in the perinatal period, is linked to an increased risk of fetal CTDs. Further longitudinal analyses are needed to validate these results.


Subject(s)
Air Pollutants , Heart Defects, Congenital , Maternal Exposure , Ozone , Ozone/toxicity , Female , Humans , Pregnancy , Maternal Exposure/adverse effects , Heart Defects, Congenital/chemically induced , Heart Defects, Congenital/epidemiology , Adult , China , Air Pollutants/toxicity , Cohort Studies , Young Adult
18.
Ecotoxicol Environ Saf ; 277: 116398, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38677066

ABSTRACT

BACKGROUND: PM2.5 and its chemical components increase health risks and are associated with depression and gut microbiota. However, there is still limited evidence on whether gut microbiota and short-chain fatty acids (SCFAs) mediate the association between PM2.5, PM2.5 chemical components, and antenatal depression. The purpose of this study was to investigate the mediating role of maternal gut microbiota in correlations between short-term exposure to PM2.5, short-term exposure to PM2.5 chemical components, and antenatal depression. METHODS: Demographic information and stool samples were collected from 75 pregnant women in their third trimester. Their exposure to PM2.5 and PM2.5 chemical components was measured. Participants were divided into the non-antenatal depression group or the antenatal depression group according to the cut-off of 10 points on the Edinburgh Postnatal Depression Scale (EPDS). The gut microbiota were analyzed using the 16 S rRNA-V3/V4 gene sequence, and the concentration of PM2.5 and its chemical components was calculated using the Tracking Air Pollution in China (TAP) database. Gas chromatography-mass spectrometry was used to analyze SCFAs in stool samples. In order to assess the mediating effects of gut microbiota and SCFAs, mediation models were utilized. RESULTS: There were significant differences between gut microbial composition and SCFAs concentrations between the non-antenatal depression group and the antenatal depression group. PM2.5 and its chemical components were positively associated with EPDS scores and negatively associated with genera Enterococcus and Enterobacter. Genera Candidatus_Soleaferrea (ß = -7.21, 95%CI -11.00 to -3.43, q = 0.01) and Enterococcus (ß = -2.37, 95%CI -3.87 to -0.87, q = 0.02) were negatively associated with EPDS scores, indicating their potential protective effects against antenatal depression. There was no significant association between SCFAs and EPDS scores. The mediating role of Enterococcus between different lagged periods of PM2.5, PM2.5 chemical component exposure, and antenatal depression was revealed. For instance, Enterococcus explained 29.23% (95%CI 2.16-87.13%, p = 0.04) of associations between PM2.5 exposure level at the day of sampling (lag 0) and EPDS scores. CONCLUSION: Our study highlights that Enterococcus may mediate the associations between PM2.5, PM2.5 chemical components, and antenatal depression. The mediating mechanism through which the gut microbiota influences PM2.5-induced depression in pregnant women still needs to be further studied.


Subject(s)
Air Pollutants , Fatty Acids, Volatile , Feces , Gastrointestinal Microbiome , Particulate Matter , Gastrointestinal Microbiome/drug effects , Female , Humans , Pregnancy , Feces/microbiology , Feces/chemistry , Particulate Matter/toxicity , Fatty Acids, Volatile/analysis , Adult , Air Pollutants/analysis , China , Depression/chemically induced , Maternal Exposure/adverse effects , Maternal Exposure/statistics & numerical data
19.
Sci Rep ; 14(1): 9947, 2024 04 30.
Article in English | MEDLINE | ID: mdl-38689029

ABSTRACT

Kawasaki disease (KD) is an acute systemic vasculitis primarily affecting young children, with an unclear etiology. We investigated the link between maternal heavy metal exposure and KD incidence in children using the Japan Environment and Children's Study, a large-scale nationwide prospective cohort with approximately 100,000 mother-child pairs. Maternal blood samples collected during the second/third trimester were analyzed for heavy metals [mercury (Hg), cadmium (Cd), lead (Pb), selenium (Se), manganese (Mn)], divided into four quartiles based on concentration levels. KD incidence within the first year of life was tracked via questionnaire. Among 85,378 mother-child pairs, 316 children (0.37%) under one year were diagnosed with KD. Compared with the lowest concentration group (Q1), the highest (Q4) showed odds ratios (95% confidence interval) for Hg, 1.29 (0.82-2.03); Cd, 0.99 (0.63-1.58); Pb, 0.84 (0.52-1.34); Se, 1.17 (0.70-1.94); Mn, 0.70 (0.44-1.11), indicating no concentration-dependent increase. Sensitivity analyses with logarithmic transformation and extended outcomes up to age 3 yielded similar results. No significant association was found between maternal heavy metal levels and KD incidence, suggesting that heavy metal exposure does not increase KD risk.


Subject(s)
Maternal Exposure , Metals, Heavy , Mucocutaneous Lymph Node Syndrome , Humans , Mucocutaneous Lymph Node Syndrome/epidemiology , Mucocutaneous Lymph Node Syndrome/chemically induced , Mucocutaneous Lymph Node Syndrome/etiology , Mucocutaneous Lymph Node Syndrome/blood , Female , Japan/epidemiology , Metals, Heavy/blood , Metals, Heavy/adverse effects , Pregnancy , Maternal Exposure/adverse effects , Male , Adult , Prospective Studies , Infant , Incidence , Prenatal Exposure Delayed Effects/epidemiology , Child, Preschool , Cadmium/blood , Cadmium/adverse effects
20.
Toxicology ; 504: 153796, 2024 May.
Article in English | MEDLINE | ID: mdl-38582279

ABSTRACT

As a broad-spectrum and efficient insecticide, beta-Cypermethrin (ß-CYP) poses a health risk to pregnancy. It matters the mechanisms of maternal exposure to ß-CYP for impacting reproductive health. The placenta, a transient organ pivotal for maternal-fetal communication during pregnancy, plays a crucial role in embryonic development. The effect of ß-CYP exposure on the placenta and its underlying molecular mechanisms remain obscure. The objective of this study was to investigate the effect of ß-CYP exposure on placental development and the function of trophoblast, as well as the underlying mechanisms through CD-1 mouse model (1, 10, 20 mg/kg.bw) and in vitro HTR-8/SVneo cell model (12.5, 25, 50, 100 µM). We found slower weight gain and reduced uterine wet weight in pregnant mice with maternal exposure to ß-CYP during pregnancy, as well as adverse pregnancy outcomes such as uterine bleeding and embryo resorption. The abnormal placental development in response to ß-CYP was noticed, including imbalanced placental structure and disrupted labyrinthine vascular development. Trophoblasts, pivotal in placental development and vascular remodeling, displayed abnormal differentiation under ß-CYP exposure. This aberration was characterized by thickened trophoblast layers in the labyrinthine zone, accompanied by mitochondrial and endoplasmic reticulum swelling within trophoblasts. Further researches on human chorionic trophoblast cell lines revealed that ß-CYP exposure induced apoptosis in HTR-8/SVneo cells. This induction resulted in a notable decrease in migration and invasion abilities, coupled with oxidative stress and the inhibition of the Notch signaling pathway. N-acetylcysteine (an antioxidant) partially restored the impaired Notch signaling pathway in HTR-8/SVneo cells, and mitigated cellular functional damage attributed to ß-CYP exposure. Collectively, exposure to ß-CYP induced oxidative stress and then led to inhibition of the Notch signaling pathway and dysfunction of trophoblast cells, ultimately resulted in abnormal placenta and pregnancy. These findings indicate Reactive Oxygen Species as potential intervention targets to mitigate ß-CYP toxicity. The comprehensive elucidation contributes to our understanding of ß-CYP biosafety and offers an experimental basis for preventing and managing its reproductive toxicity.


Subject(s)
Insecticides , Oxidative Stress , Pyrethrins , Trophoblasts , Pyrethrins/toxicity , Female , Pregnancy , Trophoblasts/drug effects , Trophoblasts/pathology , Trophoblasts/metabolism , Oxidative Stress/drug effects , Animals , Mice , Insecticides/toxicity , Humans , Maternal Exposure/adverse effects , Placentation/drug effects , Cell Line , Placenta/drug effects , Placenta/pathology , Placenta/metabolism , Apoptosis/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...