Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.348
Filter
1.
J Immunotoxicol ; 21(1): 2340495, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38946256

ABSTRACT

Per- and polyfluoroalkyl substances (PFAS) are anthropogenic organofluorine compounds that persist indefinitely in the environment and bioaccumulate throughout all trophic levels. Biomonitoring efforts have detected multiple PFAS in the serum of most people. Immune suppression has been among the most consistent effects of exposure to PFAS. PFAS often co-occur as mixtures in the environment, however, few studies have examined immunosuppression of PFAS mixtures or determined whether PFAS exposure affects immune function in the context of infection. In this study, mixtures containing two or four different PFAS and a mouse model of infection with influenza A virus (IAV) were used to assess immunotoxicity of PFAS mixtures. PFAS were administered via the drinking water as either a binary mixture of perfluorooctane sulfonate (PFOS) and perfluorooctanoic acid (PFOA) or quaternary mixture of PFOS, PFOA, perfluorohexane sulfonate (PFHxS), and perfluorononanoic acid (PFNA). The results indicated that the binary mixture affected the T-cell response, while the quaternary mixture affected the B-cell response to infection. These findings indicate that the immunomodulatory effects of PFAS mixtures are not simply additive, and that the sensitivity of immune responses to PFAS varies by cell type and mixture. The study also demonstrates the importance of studying adverse health effects of PFAS mixtures.


Subject(s)
Alkanesulfonic Acids , Caprylates , Fluorocarbons , Influenza A virus , Orthomyxoviridae Infections , Fluorocarbons/adverse effects , Fluorocarbons/toxicity , Animals , Mice , Influenza A virus/immunology , Alkanesulfonic Acids/toxicity , Alkanesulfonic Acids/adverse effects , Orthomyxoviridae Infections/immunology , Caprylates/toxicity , Caprylates/adverse effects , Humans , Female , Mice, Inbred C57BL , Influenza, Human/immunology , Disease Models, Animal , T-Lymphocytes/immunology , T-Lymphocytes/drug effects
2.
J Environ Sci (China) ; 145: 1-12, 2024 Nov.
Article in English | MEDLINE | ID: mdl-38844310

ABSTRACT

The potential association between colorectal cancer (CRC) and environmental pollutants is worrisome. Previous studies have found that some perfluoroalkyl acids, including perfluorooctane sulfonate (PFOS), induced colorectal tumors in experimental animals and promoted the migration of and invasion by CRC cells in vitro, but the underlying mechanism is unclear. Here, we investigated the effects of PFOS on the proliferation and migration of CRC cells and the potential mechanisms involving activating the PI3K/Akt-NF-κB signal pathway and epithelial-mesenchymal transition (EMT). It was found that PFOS promoted the growth and migration of HCT116 cells at non-cytotoxic concentrations and increased the mRNA expression of the migration-related angiogenic cytokines vascular endothelial growth factor (VEGF) and interleukin-8 (IL-8). In a mechanistic investigation, the up-stream signal pathway PI3K/Akt-NF-κB was activated by PFOS, and the process was suppressed by LY294002 (PI3K/Akt inhibitor) and BAY11-7082 (NF-κB inhibitor) respectively, leading to less proliferation of HCT116 cells. Furthermore, matrix metalloproteinases (MMP) and EMT-related markers were up-regulated after PFOS exposure, and were also suppressed respectively by LY294002 and BAY11-7082. Moreover, the up-regulation of EMT markers was suppressed by a MMP inhibitor GM6001. Taken together, our results indicated that PFOS promotes colorectal cancer cell migration and proliferation by activating the PI3K/Akt-NF-κB signal pathway and epithelial-mesenchymal transition. This could be a potential toxicological mechanism of PFOS-induced malignant development of colorectal cancer.


Subject(s)
Alkanesulfonic Acids , Cell Movement , Colorectal Neoplasms , Epithelial-Mesenchymal Transition , Fluorocarbons , Fluorocarbons/toxicity , Alkanesulfonic Acids/toxicity , Epithelial-Mesenchymal Transition/drug effects , Colorectal Neoplasms/pathology , Humans , Cell Movement/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , Environmental Pollutants/toxicity , HCT116 Cells , Proto-Oncogene Proteins c-akt/metabolism , NF-kappa B/metabolism , Cell Proliferation/drug effects , Cell Line, Tumor
3.
Cells ; 13(11)2024 May 30.
Article in English | MEDLINE | ID: mdl-38891072

ABSTRACT

This study explores the impact of environmental pollutants on nuclear receptors (CAR, PXR, PPARα, PPARγ, FXR, and LXR) and their heterodimerization partner, the Retinoid X Receptor (RXR). Such interaction may contribute to the onset of non-alcoholic fatty liver disease (NAFLD), which is initially characterized by steatosis and potentially progresses to steatohepatitis and fibrosis. Epidemiological studies have linked NAFLD occurrence to the exposure to environmental contaminants like PFAS. This study aims to assess the simultaneous activation of nuclear receptors via perfluorooctanoic acid (PFOA) and RXR coactivation via Tributyltin (TBT), examining their combined effects on steatogenic mechanisms. Mice were exposed to PFOA (10 mg/kg/day), TBT (5 mg/kg/day) or a combination of them for three days. Mechanisms underlying hepatic steatosis were explored by measuring nuclear receptor target gene and lipid metabolism key gene expressions, by quantifying plasma lipids and hepatic damage markers. This study elucidated the involvement of the Liver X Receptor (LXR) in the combined effect on steatosis and highlighted the permissive nature of the LXR/RXR heterodimer. Antagonistic effects of TBT on the PFOA-induced activation of the Pregnane X Receptor (PXR) and Peroxisome Proliferator-Activated Receptor Gamma (PPARγ) were also observed. Overall, this study revealed complex interactions between PFOA and TBT, shedding light on their combined impact on liver health.


Subject(s)
Caprylates , Fluorocarbons , Trialkyltin Compounds , Animals , Trialkyltin Compounds/pharmacology , Caprylates/pharmacology , Mice , Fluorocarbons/toxicity , Fluorocarbons/pharmacology , Male , Mice, Inbred C57BL , Liver X Receptors/metabolism , Liver/metabolism , Liver/drug effects , Liver/pathology , Retinoid X Receptors/metabolism , Fatty Liver/metabolism , Fatty Liver/chemically induced , Fatty Liver/pathology , Receptors, Cytoplasmic and Nuclear/metabolism , Lipid Metabolism/drug effects , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Non-alcoholic Fatty Liver Disease/chemically induced
4.
Ecotoxicol Environ Saf ; 280: 116524, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38838464

ABSTRACT

BACKGROUND: Organophosphate esters (OPEs) and Per- and polyfluoroalkyl substances (PFAS) are ubiquitous environmental contaminants with common exposure sources, leading to their widespread presence in human body. However, evidence on co-exposure to OPEs and PFAS and its impact on cardiovascular-kidney-liver-metabolic biomarkers remains limited. METHODS: In this cross-sectional study, 467 adults were enrolled from January to May 2022 during physical visits in Shijiazhuang, Hebei province. Eleven types of OPEs and twelves types of PFAS were detected, among which eight OPEs and six PFAS contaminants were detected in more than 60% of plasma samples. Seventeen biomarkers were assessed to comprehensively evaluate the cardiovascular-kidney-liver-metabolic function. Multiple linear regression, multipollutant models with sparse partial least squares, and Bayesian kernel machine regression (BKMR) models were applied to examine the associations of individual OPEs and PFAS and their mixtures with organ function and metabolism, respectively. RESULTS: Of the over 400 exposure-outcome associations tested when modelling, we observed robust results across three models that perfluorohexanoic acid (PFHxS) was significantly positively associated with alanine aminotransferase (ALT), aspartate aminotransferase (AST), total bilirubin (TBIL), and indirect bilirubin (IBIL). Perfluorononanoic acid was significantly associated with decreased AST/ALT and increased very-low-density lipoprotein cholesterol levels. Besides, perfluorodecanoic acid was correlated with increased high lipoprotein cholesterol and perfluoroundecanoic acid was consistently associated with lower glucose level. BKMR analysis showed that OPEs and PFAS mixtures were positively associated with IBIL and TBIL, among which PFHxS was the main toxic chemicals. CONCLUSIONS: Our findings suggest that exposure to OPEs and PFAS, especially PFHxS and PFNA, may disrupt organ function and metabolism in the general population, providing insight into the potential pathophysiological mechanisms of OPEs and PFAS co-exposure and chronic diseases.


Subject(s)
Biomarkers , Environmental Pollutants , Esters , Fluorocarbons , Kidney , Liver , Organophosphates , Humans , Biomarkers/blood , Female , Male , Cross-Sectional Studies , Adult , Fluorocarbons/blood , Fluorocarbons/toxicity , China , Middle Aged , Environmental Pollutants/blood , Liver/drug effects , Kidney/drug effects , Organophosphates/toxicity , Environmental Exposure/statistics & numerical data , Caproates , Young Adult , Aged , East Asian People
5.
Ecotoxicol Environ Saf ; 280: 116553, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38850699

ABSTRACT

The incidence of nonalcoholic steatohepatitis (NASH) is related with perfluorooctane sulfonate (PFOS), yet the mechanism remains ill-defined. Mounting evidence suggests that ferroptosis plays a crucial role in the initiation of NASH. In this study, we used mice and human hepatocytes L-02 to investigate the role of ferroptosis in PFOS-induced NASH and the effect and molecular mechanism of PFOS on liver ferroptosis. We found here that PFOS caused NASH in mice, and lipid accumulation and inflammatory response in the L-02 cells. PFOS induced hepatic ferroptosis in vivo and in vitro, as evidenced by the decrease in glutathione peroxidase 4 (GPX4), and the increases in cytosolic iron, acyl-CoA synthetase long-chain family member 4 (ACSL4) and lipid peroxidation. In the PFOS-treated cells, the increases in the inflammatory factors and lipid contents were reversed by ferroptosis inhibitor. PFOS-induced ferroptosis was relieved by autophagy inhibitor. The expression of mitochondrial calcium uniporter (MCU) was accelerated by PFOS, leading to subsequent mitochondrial calcium accumulation, and inhibiting autophagy reversed the increase in MCU. Inhibiting mitochondrial calcium reversed the variations in GPX4 and cytosolic iron, without influencing the change in ACSL4, induced by PFOS. MCU interacted with ACSL4 and the siRNA against MCU reversed the changes in ACSL4,GPX4 and cytosolic iron systemically. This study put forward the involvement of hepatic ferroptosis in PFOS-induced NASH and identified MCU as the mediator of the autophagy-dependent ferroptosis.


Subject(s)
Alkanesulfonic Acids , Autophagy , Calcium , Coenzyme A Ligases , Ferroptosis , Fluorocarbons , Non-alcoholic Fatty Liver Disease , Ferroptosis/drug effects , Fluorocarbons/toxicity , Animals , Alkanesulfonic Acids/toxicity , Mice , Non-alcoholic Fatty Liver Disease/chemically induced , Non-alcoholic Fatty Liver Disease/pathology , Autophagy/drug effects , Coenzyme A Ligases/metabolism , Humans , Calcium/metabolism , Calcium Channels/metabolism , Male , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Cell Line , Hepatocytes/drug effects
6.
Pharmacol Res ; 205: 107251, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38862070

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is one of the leading causes of chronic liver disease worldwide. Epidemiological studies have reported that exposure of the population to environmental endocrine-disrupting chemicals (EDCs) is associated with NAFLD. However, EDCs are of different types, and there are inconsistencies in the relevant evidence and descriptions, which have not been systematically summarized so far. Therefore, this study aimed to determine the association between population exposure to EDCs and NAFLD. Three databases, including PubMed, Web of science, and Embase were searched, and 27 articles were included in this study. Methodological quality, heterogeneity, and publication bias of the included studies were assessed using the Newcastle-Ottawa scale, I2 statistics, Begg's test, and Egger's test. The estimated effect sizes of the included studies were pooled and evaluated using the random-effects model (I2 > 50 %) and the fixed-effects model ( I2 < 50 %). The pooled-estimate effect sizes showed that population exposure to Phthalates (PAEs) (OR = 1.18, 95 % CI:1.03-1.34), cadmium (Cd) (OR = 1.37, 95 % CI:1.09-1.72), and bisphenol A (OR = 1.43, 95 % CI:1.24-1.65) were positively correlated with the risk of NAFLD. Exposure to mercury (OR =1.46, 95 % CI:1.17-1.84) and Cd increased the risk of "elevated alanine aminotransferase". On the contrary, no significant association was identified between perfluoroalkyl substances (OR =0.99, 95 % CI:0.93-1.06) and NAFLD. However, female exposure to perfluorooctanoic acid (OR =1.82, 95 % CI:1.01-3.26) led to a higher risk of NAFLD than male exposure. In conclusion, this study revealed that EDCs were risk factors for NAFLD. Nonetheless, the sensitivity analysis results of some of the meta-analyses were not stable and demonstrated high heterogeneity. The evidence for these associations is limited, and more large-scale population-based studies are required to confirm these findings.


Subject(s)
Endocrine Disruptors , Non-alcoholic Fatty Liver Disease , Non-alcoholic Fatty Liver Disease/epidemiology , Non-alcoholic Fatty Liver Disease/chemically induced , Humans , Endocrine Disruptors/adverse effects , Endocrine Disruptors/toxicity , Phthalic Acids/adverse effects , Phthalic Acids/toxicity , Environmental Pollutants/adverse effects , Environmental Pollutants/toxicity , Phenols/adverse effects , Phenols/toxicity , Benzhydryl Compounds/adverse effects , Cadmium/adverse effects , Cadmium/toxicity , Fluorocarbons/adverse effects , Fluorocarbons/toxicity
7.
J Hazard Mater ; 474: 134790, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-38850938

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC)/pancreatic cancer, is a highly aggressive malignancy with poor prognosis. Gemcitabine-based chemotherapy remains the cornerstone of PDAC treatment. Nonetheless, the development of resistance to gemcitabine among patients is a major factor contributing to unfavorable prognostic outcomes. The resistance exhibited by tumors is modulated by a constellation of factors such as genetic mutations, tumor microenvironment transforms, environmental contaminants exposure. Currently, comprehension of the relationship between environmental pollutants and tumor drug resistance remains inadequate. Our study found that PFOS/6:2 Cl-PFESA exposure increases resistance to gemcitabine in PDAC. Subsequent in vivo trials confirmed that exposure to PFOS/6:2 Cl-PFESA reduces gemcitabine's efficacy in suppressing PDAC, with the inhibition rate decreasing from 79.5 % to 56.7 %/38.7 %, respectively. Integrative multi-omics sequencing and molecular biology analyses have identified the upregulation of ribonucleotide reductase catalytic subunit M1 (RRM1) as a critical factor in gemcitabine resistance. Subsequent research has demonstrated that exposure to PFOS and 6:2 Cl-PFESA results in the upregulation of the RRM1 pathway, consequently enhancing chemotherapy resistance. Remarkably, the influence exerted by 6:2 Cl-PFESA exceeds that of PFOS. Despite 6:2 Cl-PFESA being regarded as a safer substitute for PFOS, its pronounced effect on chemotherapeutic resistance in PDAC necessitates a thorough evaluation of its potential risks related to gastrointestinal toxicity.


Subject(s)
Alkanesulfonic Acids , Carcinoma, Pancreatic Ductal , Deoxycytidine , Drug Resistance, Neoplasm , Fluorocarbons , Gemcitabine , Pancreatic Neoplasms , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Pancreatic Neoplasms/drug therapy , Humans , Fluorocarbons/toxicity , Alkanesulfonic Acids/toxicity , Cell Line, Tumor , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Drug Resistance, Neoplasm/drug effects , Animals , Ribonucleoside Diphosphate Reductase , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Antimetabolites, Antineoplastic/therapeutic use , Female , Mice , Male , Mice, Nude
8.
Sci Total Environ ; 940: 173562, 2024 Aug 25.
Article in English | MEDLINE | ID: mdl-38825197

ABSTRACT

Epidemic and animal studies have reported that perfluoroalkyl and polyfluoroalkyl substances (PFASs) are strongly associated with liver injury; however, to date, the effects of PFASs on the hepatic microenvironment remain largely unknown. In this study, we established perfluorooctane sulfonic acid (PFOS)-induced liver injury models by providing male and female C57BL/6 mice with water containing PFOS at varying doses for 4 weeks. Hematoxylin and eosin staining revealed that PFOS induced liver injury in both sexes. Elevated levels of serum aminotransferases including those of alanine aminotransferase and aspartate transaminase were detected in the serum of mice treated with PFOS. Female mice exhibited more severe liver injury than male mice. We collected the livers from female mice and performed single-cell RNA sequencing. In total, 36,529 cells were included and grouped into 10 major cell types: B cells, granulocytes, T cells, NK cells, monocytes, dendritic cells, macrophages, endothelial cells, fibroblasts, and hepatocytes. Osteoclast differentiation was upregulated and the T cell receptor signaling pathway was significantly downregulated in PFOS-treated livers. Further analyses revealed that among immune cell clusters in PFOS-treated livers, Tcf7+CD4+T cells were predominantly downregulated, whereas conventional dendritic cells and macrophages were upregulated. Among the fibroblast subpopulations, hepatic stellate cells were significantly enriched in PFOS-treated female mice. CellphoneDB analysis suggested that fibroblasts interact closely with endothelial cells. The major ligand-receptor pairs between fibroblasts and endothelial cells in PFOS-treated livers were Dpp4_Cxcl12, Ackr3_Cxcl12, and Flt1_complex_Vegfa. These genes are associated with directing cell migration and angiogenesis. Our study provides a general framework for understanding the microenvironment in the livers of female mice exposed to PFOS at the single-cell level.


Subject(s)
Alkanesulfonic Acids , Fluorocarbons , Mice, Inbred C57BL , Animals , Fluorocarbons/toxicity , Alkanesulfonic Acids/toxicity , Female , Mice , Male , Chemical and Drug Induced Liver Injury/genetics , Transcriptome/drug effects , Liver/drug effects , Single-Cell Analysis , Environmental Pollutants/toxicity
9.
Expert Rev Endocrinol Metab ; 19(4): 307-315, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38764236

ABSTRACT

INTRODUCTION: The environmental spread of pollutants has led to a persistent exposure of living beings to multiple chemicals, by now become ubiquitous in the surrounding environment. Environmental exposure to these substances has been reported to cause multi- and/or transgenerational health effects. Per- and Polyfluorinated Substances (PFAS) raise great concern, given their known effects both as endocrine disruptors and potential carcinogens. The multi/trans-generational effects of different endocrine disruptors have been investigated by several studies, and harmful effects observed also for PFAS. AREAS COVERED: This review examines the current data on the multi-trans-generational effects of PFAS, with a focus on their impact on the thyroid axis. The aim is to determine if there is evidence of potential multi-trans-generational effects of PFAS on the thyroid and/or if more research is needed. EXPERT OPINION: PFAS exposure impacts thyroid homeostasis and can cross the placental barrier. In addition PFAS have shown multi-transgenerational effects in laboratory experiences and animal models, but thyroid disruptive effects of PFAS were also investigated only in a small number of these studies. Efforts are needed to study the adverse effects of PFAS, as not all PFAS are regulated and removal strategies are still being developed.


Subject(s)
Endocrine Disruptors , Environmental Pollutants , Fluorocarbons , Thyroid Gland , Humans , Endocrine Disruptors/adverse effects , Thyroid Gland/drug effects , Animals , Fluorocarbons/adverse effects , Fluorocarbons/toxicity , Female , Environmental Pollutants/toxicity , Pregnancy , Environmental Exposure/adverse effects , Prenatal Exposure Delayed Effects/chemically induced
10.
Environ Toxicol Chem ; 43(7): 1638-1647, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38721889

ABSTRACT

The European honey bee (Apis mellifera) is an important crop pollinator threatened by multiple stressors, including exposure to contaminants. Perfluorooctane sulfonate (PFOS) is a persistent global contaminant that accumulates and biomagnifies in food chains and is detected in honey. Even sublethal exposure to PFOS is detrimental to bee health, but exposure routes are unclear and nothing is known about bee response (detection, avoidance, or attraction) to PFOS. Using Y-mazes, we studied the response of individual bees to PFOS-spiked sugar syrup at four concentrations, 0.02, 30, 61 and 103 µg L-1. Bee activity, choice behavior, and drink duration for unspiked and spiked sugar syrup was recorded for 10 min in the Y-maze system. Most bees (≥80%) tasted and then drank the sugar syrup solutions, including the PFOS-contaminated syrup. Only at 61 and 103 µg L-1 did bees significantly avoid drinking PFOS-spiked syrup, and only when given a choice with unspiked syrup. When the choice of consuming unspiked syrup was removed, the bees drank PFOS-spiked syrup at all the PFOS concentrations tested, and avoidance was not evident. Avoidance was not observed in any treatment at 0.02 or 30 µg L-1 PFOS, concentrations that are frequently reported in environmental waters in contaminated areas. These findings confirm that bees will access PFOS-contaminated resources at concentrations detrimental to the colony health, and provide evidence of potential exposure pathways that may threaten crop pollination services and also human health via food chain transfer in PFOS-contaminated areas. Environ Toxicol Chem 2024;43:1638-1647. © 2024 SETAC.


Subject(s)
Alkanesulfonic Acids , Fluorocarbons , Bees/drug effects , Fluorocarbons/toxicity , Fluorocarbons/analysis , Alkanesulfonic Acids/toxicity , Animals , Environmental Pollutants/toxicity , Environmental Pollutants/analysis , Behavior, Animal/drug effects
11.
Mar Pollut Bull ; 203: 116446, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38703627

ABSTRACT

Perfluorooctanesulfonic acid (PFOS) is detected in estuarine environments, where salinity levels fluctuate regularly. We investigated the effects of salinity on the toxicity of PFOS in embryos and larvae of Cyprinodon variegatus. We crossed six PFOS treatments (0, 1-10,000 µg/L) with two salinities (10, 30 ppt). Larvae exposed to the highest concentration of PFOS under high salinity accumulated over twice the amount of PFOS compared to larvae maintained under low salinity. Embryonic survival was unaffected by PFOS, salinity, or their interaction. PFOS delayed time to hatch and increased salinity reduced time to hatch regardless of PFOS treatment; however, no salinity by PFOS interactions were observed. Conversely, PFOS and salinity interacted in the larval stage, with decreased survival at 30 ppt salinity. This is one of the first studies evaluating interactive effects of PFOS and high salinity and highlights the importance of assessing PFAS toxicity across life stages.


Subject(s)
Alkanesulfonic Acids , Fluorocarbons , Larva , Salinity , Water Pollutants, Chemical , Animals , Fluorocarbons/toxicity , Alkanesulfonic Acids/toxicity , Water Pollutants, Chemical/toxicity , Larva/drug effects , Estuaries , Killifishes/physiology , Embryo, Nonmammalian/drug effects
12.
Environ Res ; 256: 119221, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38795951

ABSTRACT

Per- and polyfluoroalkyl substances (PFAS) constitutes a group of highly persistent man-made substances. Recent evidence indicates that PFAS negatively impact the immune system. However, it remains unclear how different PFAS are associated with alterations in circulating leukocyte subpopulations. More detailed knowledge of such potential associations can provide better understanding into mechanisms of PFAS immunotoxicity in humans. In this exploratory study, associations of serum levels of common PFAS (perfluorooctanoic acid (PFOA), perfluorooctane sulfonic acid (PFOS), perfluorononanoic acid (PFNA), and perfluorohexane sulfonic acid (PFHxS)) and immune cell profiles of peripheral blood mononuclear cells, both with and without immunostimulation, were investigated. High-dimensional single cell analysis by mass cytometry was done on blood leukocytes from fifty participants in the Norwegian human biomonitoring EuroMix study. Different PFAS were associated with changes in various subpopulations of natural killer (NK), T helper (Th), and cytotoxic T (Tc) cells. Broadly, PFAS concentrations were related to increased frequencies of NK cells and activated subpopulations of NK cells. Additionally, increased levels of activated T helper memory cell subpopulations point to Th2/Th17 and Treg-like skewed profiles. Finally, PFAS concentrations were associated with decreased frequencies of T cytotoxic cell subpopulations with CXCR3+ effector memory (EM) phenotypes. Several of these observations point to biologically plausible mechanisms that may contribute to explaining the epidemiological reports of immunosuppression by PFAS. Our results suggest that PFAS exposures even at relatively low levels are associated with changes in immune cell subpopulations, a finding which should be explored more thoroughly in a larger cohort. Additionally, causal relationships should be confirmed in experimental studies. Overall, this study demonstrates the strength of profiling by mass cytometry in revealing detailed changes in immune cells at a single cell level.


Subject(s)
Fluorocarbons , Killer Cells, Natural , Humans , Fluorocarbons/toxicity , Fluorocarbons/blood , Male , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Adult , Female , Middle Aged , Environmental Pollutants/toxicity , Environmental Pollutants/blood , Environmental Exposure , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology , Norway , Alkanesulfonic Acids/toxicity , Alkanesulfonic Acids/blood , Aged
13.
Chemosphere ; 359: 142332, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38754493

ABSTRACT

Perfluorooctanesulfonic acid (PFOS) is a widely recognized environment pollutant known for its high bioaccumulation potential and a long elimination half-life. Several studies have shown that PFOS can alter multiple biological pathways and negatively affect human health. Considering the direct exposure to the gastrointestinal (GI) tract to environmental pollutants, PFOS can potentially disrupt intestinal homeostasis. However, there is limited knowledge about the effect of PFOS exposure on normal intestinal tissues, and its contribution to GI-associated diseases remains to be determined. In this study, we examined the effect of PFOS exposure on the gene expression profile of intestinal tissues of C57BL/6 mice using RNAseq analysis. We found that PFOS exposure in drinking water significantly downregulates mitochondrial 3-hydroxy-3-methylglutaryl-CoA synthase 2 (HMGCS2), a rate-limiting ketogenic enzyme, in intestinal tissues of mice. We found that diets containing the soluble fibers inulin and pectin, which are known to be protective against PFOS exposure, were ineffective in reversing the downregulation of HMGCS2 expression in vivo. Analysis of intestinal tissues also demonstrated that PFOS exposure leads to upregulation of proteins implicated in colorectal carcinogenesis, including ß-catenin, c-MYC, mTOR and FASN. Consistent with the in vivo results, PFOS exposure leads to downregulation of HMGCS2 in mouse and human normal intestinal organoids in vitro. Furthermore, we show that shRNA-mediated knockdown of HMGCS2 in a human normal intestinal cell line resulted in increased cell proliferation and upregulation of key proliferation-associated proteins such as cyclin D, survivin, ERK1/2 and AKT, along with an increase in lipid accumulation. In summary, our results suggest that PFOS exposure may contribute to pathological changes in normal intestinal cells via downregulation of HMGCS2 expression and upregulation of pro-carcinogenic signaling pathways that may increase the risk of colorectal cancer development.


Subject(s)
Alkanesulfonic Acids , Carcinogenesis , Down-Regulation , Fluorocarbons , Hydroxymethylglutaryl-CoA Synthase , Mice, Inbred C57BL , Animals , Alkanesulfonic Acids/toxicity , Fluorocarbons/toxicity , Hydroxymethylglutaryl-CoA Synthase/metabolism , Hydroxymethylglutaryl-CoA Synthase/genetics , Mice , Down-Regulation/drug effects , Intestinal Neoplasms/chemically induced , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Up-Regulation/drug effects , Environmental Pollutants/toxicity , Intestines/drug effects , Humans , Intestinal Mucosa/metabolism
14.
J Hazard Mater ; 474: 134683, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-38820745

ABSTRACT

The microbial community present in our intestines is pivotal for converting indigestible substances into vital nutrients and signaling molecules such as short-chain fatty acids (SCFAs). These compounds have considerable influence over our immune system and the development of diverse human diseases. However, ingested environmental contaminants, known as xenobiotics, can upset the delicate balance of the microbial gut community and enzymatic processes, consequently affecting the host organism. In our study, we employed an in vitro bioreactor model system based on the simplified human microbiome model (SIHUMIx) to investigate the direct effects of specific xenobiotics, such as perfluorooctanoic acid (PFOA), perfluorohexanoic acid (PFHxA) and perfluorobutanoic acid (PFBA) or bisphenol S (BPS) and bisphenol F (BPF), either individually or in combination, on the microbiota. We observed increased SCFA production, particularly acetate and butyrate, with PFAS exposure. Metaproteomics revealed pathway alterations across treatments, including changes in vitamin synthesis and fatty acid metabolism with BPX. This study underscores the necessity of assessing the combined effects of xenobiotics to better safeguard public health. It emphasizes the significance of considering adverse effects on the microbiome in the risk assessment of environmental chemicals.


Subject(s)
Benzhydryl Compounds , Fatty Acids, Volatile , Fluorocarbons , Gastrointestinal Microbiome , Xenobiotics , Humans , Xenobiotics/toxicity , Xenobiotics/metabolism , Fluorocarbons/toxicity , Gastrointestinal Microbiome/drug effects , Fatty Acids, Volatile/metabolism , Benzhydryl Compounds/toxicity , Phenols/toxicity , Bioreactors , Sulfones/toxicity , Environmental Pollutants/toxicity
15.
Ecotoxicol Environ Saf ; 279: 116453, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38772139

ABSTRACT

Chlorinated polyfluorinated ether sulfonate, commercially known as F-53B, has been associated with adverse birth outcomes. However, the reproductive toxicology of F-53B on the placenta remains poorly understood. To address this gap, we examined the impact of F-53B on placental injury and its underlying molecular mechanisms in vivo. Pregnant C57BL/6 J female mice were randomly allocated to three groups: the control group, F-53B 0.8 µg/kg/day group, and F-53B 8 µg/kg/day group. After F-53B exposure through free drinking water from gestational day (GD) 0.5-14.5, the F-53B 8 µg/kg/day group exhibited significant increases in placental weights and distinctive histopathological alterations, including inflammatory cell infiltration, heightened syncytiotrophoblast knots, and a loosened trophoblastic basement membrane. Within the F-53B 8 µg/kg/day group, placental tissue exhibited increased apoptosis, as indicated by increased caspase3 activation. Furthermore, F-53B potentially induced the NF-κB signaling pathway activation through IκB-α phosphorylation. Subsequently, this activation upregulated the expression of inflammatory cytokines and components of the NLRP3 inflammasome, including activated caspase1, IL-1ß, IL-18, and cleaved gasdermin D (GSDMD), ultimately leading to pyroptosis in the mouse placenta. Our findings reveal a pronounced inflammatory injury in the placenta due to F-53B exposure, suggesting potential reproductive toxicity at concentrations relevant to the human population. Further toxicological and epidemiological investigations are warranted to conclusively assess the reproductive health risks posed by F-53B.


Subject(s)
Inflammasomes , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein , Placenta , Animals , Female , Pregnancy , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Placenta/drug effects , Placenta/pathology , Mice , Inflammasomes/drug effects , Inflammation/chemically induced , Inflammation/pathology , Apoptosis/drug effects , NF-kappa B/metabolism , Fluorocarbons/toxicity , Signal Transduction/drug effects
16.
J Hazard Mater ; 473: 134642, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38776814

ABSTRACT

Per- and poly-fluoroalkyl substances (PFAS) are synthetic chemicals widely used in commercial products. PFAS are a global concern due to their persistence in the environment and extensive associations with adverse health outcomes. While legacy PFAS have been extensively studied, many non-legacy PFAS lack sufficient toxicity information. In this study, we first analyzed the bioactivity of PFAS using Tox21 screening data surveying more than 75 assay endpoints (e.g., nuclear receptors, stress response, and metabolism) to understand the toxicity of non-legacy PFAS and investigate potential new targets of PFAS. From the Tox21 screening data analysis, we confirmed several known PFAS targets/pathways and identified several potential novel targets/pathways of PFAS. To confirm the effect of PFAS on these novel targets/pathways, we conducted several cell- and enzyme-based assays in the follow-up studies. We found PFAS inhibited cytochromes P450s (CYPs), especially CYP2C9 with IC50 values of < 1 µM. Considering PFAS affected other targets/pathways at > 10 µM, PFAS have a higher affinity to CYP2C9. This PFAS-CYP2C9 interaction was further investigated using molecular docking analysis. The result suggested that PFAS directly bind to the active sites of CYP2C9. These findings have important implications to understand the mechanism of PFAS action and toxicity.


Subject(s)
Cytochrome P-450 Enzyme System , Fluorocarbons , Receptors, Cytoplasmic and Nuclear , Fluorocarbons/toxicity , Cytochrome P-450 Enzyme System/metabolism , Humans , Receptors, Cytoplasmic and Nuclear/metabolism , Stress, Physiological/drug effects , Environmental Pollutants/toxicity , Molecular Docking Simulation
17.
Ecotoxicol Environ Saf ; 279: 116501, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38805831

ABSTRACT

6:2 Chlorinated polyfluoroalkyl ether sulfonate (F-53B) is a new type of perfluorinated and polyfluoroalkyl substance (PFAS) that is used extensively in industry and manufacturing. F-53B causes damage to multiple mammalian organs. However, the impacts of F-53B on bone are unknown. Maternal exposure to F-53B is of particular concern because of the vulnerability of the developing fetus and newborn to contaminants from the mother. The goal of this study was to examine the impacts of maternal F-53B exposure on bone growth and development in offspring and to explore its underlying mechanisms. Herein, C57BL/6 J mice were given free access to deionized water containing 0, 0.57, or 5.7 mg/L F-53B during pregnancy and lactation. F-53B exposure resulted in impaired liver function, decreased IGF-1 secretion, dysregulation of bone metabolism and disruption of the dynamic balance between osteoblasts and osteoclasts in male offspring. F-53B inhibits longitudinal bone growth and development and causes osteoporosis in male offspring. F-53B may affect the growth and development of offspring bone via the IGF-1/OPG/RANKL/CTSK signaling pathway. This study provides new insights for the study of short stature and bone injury caused by F-53B.


Subject(s)
Bone Development , Lactation , Maternal Exposure , Mice, Inbred C57BL , Prenatal Exposure Delayed Effects , Animals , Female , Male , Pregnancy , Mice , Maternal Exposure/adverse effects , Bone Development/drug effects , Insulin-Like Growth Factor I/metabolism , Fluorocarbons/toxicity , Osteoprotegerin/metabolism , Osteoclasts/drug effects , Bone and Bones/drug effects , Osteoblasts/drug effects , Sulfonic Acids/toxicity
18.
Chemosphere ; 361: 142446, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38801907

ABSTRACT

The extensive use of poly- and per-fluoroalkyl substances (PFASs) has les to their widespread presence in the environment, raising concerns about potential toxicity. While certain PFASs of concern have been phased-out or banned, new PFASs continue to be produced. Two such substances are perfluoroethylcyclohexane sulphonate (PFECHS) and perfluorobutane sulphamide (FBSA), replacements of perfluoroctanesulphonic acid (PFOS) that have recently been detected in multiple environmental media around the globe. Despite PFASs generally occurring in the environment as mixtures, few data are available outlining the effects of PFAS mixtures. Therefore, this research investigated the interaction potential of binary and ternary mixtures of emerging and legacy PFASs. The immortalized rainbow trout gill cell line (RTgill-W1) was chosen as the experimental model to investigate two apical endpoints: cytotoxicity and phospholipidosis. RTgill-W1 cells were exposed for 24 h to each compound to obtain endpoint-specific effect concentrations (LCx; ECx). These values were then applied to formulate mixture predictions following the Loewes Additivity and Steel and Peckham methods. Based on cytotoxicity, relative potencies of individual compounds were: PFOS > PFECHS > FSBA. PFOS and PFECHS had nearly identical effects on phospholipidosis, while FSBA did not have any effects. Most mixtures had a synergistic effect on cytotoxicity, but the effect was both dose- and ratio-dependent. PFOS and PFECHS were additive at lower concentrations (LC10) and synergistic at higher concentrations (LC50; 3:1, 1:1, and 1:3). PFECHS and FSBA mixtures were synergistic at all doses and ratios (3:1, 1:1, 1:3), while FBSA and PFOS were mainly synergistic at higher concentrations and at ratios favouring PFOS (1:1, 1:3). Tertiary combinations were mainly synergistic. For phospholipidosis, mixtures were strictly additive. These results are strongly suggestive of synergism between emerging PFAS replacements and highlight that independent apical mechanisms of different PFASs could combine to induce unexpected toxicity. Considering that emerging replacements are continuing to increase in concentration in the environment, such mixture scenarios are also likely to continue to increase in probability.


Subject(s)
Fluorocarbons , Oncorhynchus mykiss , Water Pollutants, Chemical , Fluorocarbons/toxicity , Animals , Cell Line , Water Pollutants, Chemical/toxicity , Alkanesulfonic Acids/toxicity , Drug Synergism , Sulfonic Acids
19.
Chemosphere ; 361: 142461, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38810808

ABSTRACT

With the restricted use of perfluorooctane sulfonate (PFOS) and perfluorooctanoic acid (PFOA), a number of alternatives to PFOS and PFOA have attracted great interest. Most of the alternatives are still characterized by persistence, bioaccumulation, and a variety of toxicity. Due to the production and use of these substances, they can be detected in the atmosphere, soil and water body. They affect human health through several exposure pathways and especially enter the gut by drinking water and eating food, which results in gut toxicity. In this review, we summarized the effects of PFOS, PFOA and 9 alternatives on pathological changes in the gut, the disruption of physical, chemical, biological and immune barriers of the intestine, and the gut-organ axis. This review provides a valuable understanding of the gut toxicity of PFOS, PFOA and their alternatives as well as the human health risks of emerging contaminants.


Subject(s)
Alkanesulfonic Acids , Caprylates , Environmental Pollutants , Fluorocarbons , Fluorocarbons/toxicity , Caprylates/toxicity , Alkanesulfonic Acids/toxicity , Humans , Animals , Environmental Pollutants/toxicity , Intestines/drug effects
20.
Chemosphere ; 361: 142462, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38815816

ABSTRACT

As perfluorooctanoic acid (PFOA) alternatives, hexafluoropropylene oxide dimeric acid (HFPO-DA) and hexafluoropropylene oxide trimeric acid (HFPO-TA) have been increasingly used and caused considerable water pollution. However, their toxicities to aquatic organisms are still not well known. Therefore, in this study, zebrafish embryos were exposed to PFOA (0, 1.5, 3 and 6 mg/L), HFPO-DA (0, 3, 6 and 12 mg/L) and HFPO-TA (0, 1, 2 and 4 mg/L) to comparatively investigate their thyroid disrupting effects and the developmental toxicity. Results demonstrated that waterborne exposure to PFOA and its two alternatives decreased T4 contents, the heart rate and swirl-escape rate of zebrafish embryos/larvae. The transcription levels of genes related to thyroid hormone regulation (crh), biosynthesis (tpo and tg), function (trα and trß), transport (transthyretin, ttr), and metabolism (dio1, dio2 and ugt1ab), were differently altered after the exposures, which induced the thyroid disrupting effects and decreased the heart rate. In addition, the transcription levels of some genes related to the nervous system development were also significantly affected, which was associated with the thyroid disrupting effects and consequently affected the locomotor activity of zebrafish. Therefore, HFPO-DA and HFPO-TA could not be safe alternatives to PFOA. Further studies to uncover the underlying mechanisms of these adverse effects are warranted.


Subject(s)
Embryo, Nonmammalian , Fluorocarbons , Thyroid Gland , Water Pollutants, Chemical , Zebrafish , Animals , Zebrafish/embryology , Fluorocarbons/toxicity , Thyroid Gland/drug effects , Embryo, Nonmammalian/drug effects , Water Pollutants, Chemical/toxicity , Caprylates/toxicity , Endocrine Disruptors/toxicity , Larva/drug effects , Larva/growth & development , Thyroid Hormones/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...