Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 1.003
1.
J Environ Sci (China) ; 145: 1-12, 2024 Nov.
Article En | MEDLINE | ID: mdl-38844310

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.


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
2.
Sci Total Environ ; 931: 172962, 2024 Jun 25.
Article En | MEDLINE | ID: mdl-38705306

Perfluorooctane sulfonate (PFOS) is a typical persistent organic pollutant that is characterized by environmental persistence, bioaccumulation, and toxicity. In this study, we investigated the gut microbial response of the red claw crayfish Cherax quadricarinatus after 28 days of exposure to 0 ng/L, 1 ng/L, 10 µg/L, or 10 mg/L of PFOS as a stressor. We measured oxidative stress-related enzyme activities and expression of molecules related to detoxification mechanisms to evaluate the toxic effects of PFOS. We found that PFOS disturbed microbial homeostasis in the gut of C. quadricarinatus, resulting in increased abundance of the pathogen Shewanella and decreased abundance of the beneficial bacterium Lactobacillus. The latter especially disturbed amino acid transport and carbohydrate transport. We also found that the activities of glutathione S-transferase and glutathione peroxidase were positively correlated with the expression levels of cytochrome P450 genes (GST1-1, GSTP, GSTK1, HPGDS, UGT5), which are products of PFOS-induced oxidative stress and play an antioxidant role in the body. The results of this study provided valuable ecotoxicological data to better understand the biological fate and effects of PFOS in C. quadricarinatus.


Alkanesulfonic Acids , Antioxidants , Astacoidea , Fluorocarbons , Gastrointestinal Microbiome , Oxidative Stress , Water Pollutants, Chemical , Animals , Astacoidea/drug effects , Astacoidea/physiology , Astacoidea/microbiology , Alkanesulfonic Acids/toxicity , Fluorocarbons/toxicity , Gastrointestinal Microbiome/drug effects , Water Pollutants, Chemical/toxicity , Antioxidants/metabolism , Glutathione Transferase/metabolism
3.
Environ Int ; 187: 108710, 2024 May.
Article En | MEDLINE | ID: mdl-38701644

Exposure to persistent organic pollutants (POPs), such as dichlorodiphenyltrichloroethane (DDT) and polychlorinated biphenyls (PCBs), has historically been linked to population collapses in wildlife. Despite international regulations, these legacy chemicals are still currently detected in women of reproductive age, and their levels correlate with reduced ovarian reserve, longer time-to-pregnancy, and higher risk of infertility. However, the specific modes of action underlying these associations remain unclear. Here, we examined the effects of five commonly occurring POPs - hexachlorobenzene (HCB), p,p'-dichlorodiphenyldichloroethylene (DDE), 2,3,3',4,4',5-hexachlorobiphenyl (PCB156), 2,2',3,4,4',5,5'-heptachlorobiphenyl (PCB180), perfluorooctane sulfonate (PFOS) - and their mixture on human ovaries in vitro. We exposed human ovarian cancer cell lines COV434, KGN, and PA1 as well as primary ovarian cells for 24 h, and ovarian tissue containing unilaminar follicles for 6 days. RNA-sequencing of samples exposed to concentrations covering epidemiologically relevant levels revealed significant gene expression changes related to central energy metabolism in the exposed cells, indicating glycolysis, oxidative phosphorylation, fatty acid metabolism, and reactive oxygen species as potential shared targets of POP exposures in ovarian cells. Alpha-enolase (ENO1), lactate dehydrogenase A (LDHA), cytochrome C oxidase subunit 4I1 (COX4I1), ATP synthase F1 subunit alpha (ATP5A), and glutathione peroxidase 4 (GPX4) were validated as targets through qPCR in additional cell culture experiments in KGN. In ovarian tissue cultures, we observed significant effects of exposure on follicle growth and atresia as well as protein expression. All POP exposures, except PCB180, decreased unilaminar follicle proportion and increased follicle atresia. Immunostaining confirmed altered expression of LDHA, ATP5A, and GPX4 in the exposed tissues. Moreover, POP exposures modified ATP production in KGN and tissue culture. In conclusion, our results demonstrate the disruption of cellular energy metabolism as a novel mode of action underlying POP-mediated interference of follicle growth in human ovaries.


Energy Metabolism , Fluorocarbons , Ovary , Persistent Organic Pollutants , Humans , Female , Ovary/drug effects , Ovary/metabolism , Energy Metabolism/drug effects , Fluorocarbons/toxicity , Homeostasis/drug effects , Cell Line, Tumor , Polychlorinated Biphenyls/toxicity , Dichlorodiphenyl Dichloroethylene/toxicity , Alkanesulfonic Acids/toxicity , Hexachlorobenzene/toxicity
4.
Environ Int ; 187: 108720, 2024 May.
Article En | MEDLINE | ID: mdl-38718676

BACKGROUND: Prenatal exposure to per- and polyfluoroalkyl substances (PFASs) influences neurodevelopment. Thyroid homeostasis disruption is thought to be a possible underlying mechanism. However, current epidemiological evidence remains inconclusive. OBJECTIVES: This study aimed to explore the effects of prenatal PFAS exposure on the intelligence quotient (IQ) of school-aged children and assess the potential mediating role of fetal thyroid function. METHODS: The study included 327 7-year-old children from the Sheyang Mini Birth Cohort Study (SMBCS). Cord serum samples were analyzed for 12 PFAS concentrations and 5 thyroid hormone (TH) levels. IQ was assessed using the Wechsler Intelligence Scale for Children-Chinese Revised (WISC-CR). Generalized linear models (GLM) and Bayesian Kernel Machine Regression (BKMR) were used to evaluate the individual and combined effects of prenatal PFAS exposure on IQ. Additionally, the impact on fetal thyroid function was examined using a GLM, and a mediation analysis was conducted to explore the potential mediating roles of this function. RESULTS: The molar sum concentration of perfluorinated carboxylic acids (ΣPFCA) in cord serum was significantly negatively associated with the performance IQ (PIQ) of 7-year-old children (ß = -6.21, 95 % confidence interval [CI]: -12.21, -0.21), with more pronounced associations observed among girls (ß = -9.57, 95 % CI: -18.33, -0.81) than in boys. Negative, albeit non-significant, cumulative effects were noted when considering PFAS mixture exposure. Prenatal exposure to perfluorooctanoic acid, perfluorononanoic acid, and perfluorooctanesulfonic acid was positively associated with the total thyroxine/triiodothyronine ratio. However, no evidence supported the mediating role of thyroid function in the link between PFAS exposure and IQ. CONCLUSIONS: Increased prenatal exposure to PFASs negatively affected the IQ of school-aged children, whereas fetal thyroid function did not serve as a mediator in this relationship.


Environmental Pollutants , Fluorocarbons , Intelligence , Prenatal Exposure Delayed Effects , Thyroid Gland , Humans , Female , Prenatal Exposure Delayed Effects/chemically induced , Child , Pregnancy , Fluorocarbons/toxicity , Fluorocarbons/blood , Male , Intelligence/drug effects , Thyroid Gland/drug effects , Environmental Pollutants/blood , Environmental Pollutants/toxicity , Birth Cohort , Cohort Studies , Thyroid Hormones/blood , Intelligence Tests , China , Maternal Exposure/adverse effects , Fetal Blood/chemistry , Alkanesulfonic Acids/blood , Alkanesulfonic Acids/toxicity
5.
Chemosphere ; 359: 142332, 2024 Jul.
Article En | MEDLINE | ID: mdl-38754493

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.


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
6.
Mar Environ Res ; 198: 106535, 2024 Jun.
Article En | MEDLINE | ID: mdl-38704932

One of the most difficult-to-manage new contaminants constantly released into the environment is linear alkylbenzene sulphonate (LAS), an anionic surfactant. Significant volumes of LAS are received by the Mediterranean coast of Egypt. The current study is a comprehensive assessment of the environmental fate of the LAS 1505 km off the Mediterranean coast of Egypt in the fall of 2023 in order to track its geographic spread and eventual demise in the water column. Critical analysis of LAS revealed that it is vertically distributed in various ways according to sources, uses, production amounts, and salinity levels. The vertical variation of LAS can be explained by its amphiphilic structure. A significant increase in surfactant concentration (>300 µg/L) was recorded in 66% and 43% of the total samples, ranging from 301.128 to 455.36 and from 304.556 to 486.135 for the western and eastern sides along the Egyptian Mediterranean coast, respectively. Evaluation of the average acute and chronic risk quotient (RQ) along the investigated locations revealed that fish were the most susceptible to LAS in both long and short exposure periods. The presented results also indicated significant LAS toxicity to three trophic levels (RQ values > 1). LAS toxicity to marine organisms was greater in the western than in eastern coastal regions according to acute and chronic mixture risk characterization ratios (RCRmix). The three trophic levels in the study area had the following order of acute relative contribution (RC) to LAS toxicity: fish > invertebrates > algae. The ANOVA test results showed that in both the western and eastern regions, LAS varied significantly (p < 0.05) with salinity (1.04E-60 and 5.44E-42) and depth (6.02E-65 and 1.59E-47), respectively. In addition, a significant difference was observed using the ANOVA test between the eastern and western regions of the Egyptian Mediterranean coast.


Environmental Monitoring , Surface-Active Agents , Water Pollutants, Chemical , Egypt , Water Pollutants, Chemical/toxicity , Water Pollutants, Chemical/analysis , Surface-Active Agents/toxicity , Mediterranean Sea , Animals , Alkanesulfonic Acids/toxicity , Fishes , Risk Assessment , Aquatic Organisms/drug effects , Seawater/chemistry
7.
Arch Environ Contam Toxicol ; 86(4): 383-392, 2024 May.
Article En | MEDLINE | ID: mdl-38795151

The larval fathead minnow, Pimephales promelas, 7-day subchronic survival and growth standard toxicity test method is commonly used for research and regulatory testing of effluents and compounds, including emerging contaminants such as Perfluorooctanesulfonic Acid (PFOS). Existing feeding guidelines for testing are described in multiple methods but are open to interpretation. The current study sought to determine the impact of feeding ration on P. promelas survival and biomass during a subchronic exposure to PFOS. The study was conducted in two phases: (1) a control experiment to determine the most significant feeding ration factors that maximize biomass, with consideration to laboratory logistics, and (2) application of down-selected feeding rations in a PFOS exposure to determine toxicity reference values. The control optimization study supported that feeding ration and feeding frequency were significant factors in fish biomass. In the subsequent PFOS study, fish were fed a high or low ration of Artemia twice daily, while exposed to 0.3 to 3.4 mg/L PFOS. Fish fed a high ration of Artemia had significantly (p < 0.05) greater biomass than fish fed a low ration in all exposure concentrations except 3.4 mg/L, where survival was low in both treatments. The feeding ration was not a significant factor on the survival endpoint for either treatment, but the PFOS concentration was (p < 0.0001) (high ration LC50 = 2.44 mg/L; low ration LC50 = 2.25 mg/L). These findings contribute to a better understanding of the impact feeding ration has in toxicity assessments and downstream regulatory decisions.


Alkanesulfonic Acids , Cyprinidae , Fluorocarbons , Larva , Water Pollutants, Chemical , Animals , Alkanesulfonic Acids/toxicity , Fluorocarbons/toxicity , Cyprinidae/physiology , Water Pollutants, Chemical/toxicity , Larva/drug effects , Larva/growth & development , Toxicity Tests, Subchronic
8.
Ecotoxicol Environ Saf ; 279: 116482, 2024 Jul 01.
Article En | MEDLINE | ID: mdl-38772142

Heavy metals and per- and polyfluoroalkyl substances (PFASs) have become particularly important when studying the development of depression, a common illness that severely restricts psychosocial functioning and diminishes quality of life. Therefore, the potential joint effects of heavy metal and PFAS exposure on depression, as well as the underlying mechanisms involved, were investigated by using integrated epidemiological and bioinformatic approaches in the present study. A thorough analysis of 7301 samples from the National Health and Nutrition Examination Survey (NHANES) cycles that occurred between 2005 and 2018 was performed. Single-exposure studies have shown that cadmium exposure is positively associated with depression, whereas perfluorooctanesulfonic acid (PFOS) exposure and perfluorodecanoic acid (PFDE) exposure are negatively associated with depression. Furthermore, the Bayesian kernel machine regression (BKMR) and quantile g-computation (QGcomp) models were employed to investigate the collective impact of exposure to mixed metals on depression. Cadmium emerged as the principal contributor to depression. Moreover, the addition of PFAS to the metal mixture had an antagonistic effect on depression, with PFOS having the most prominent influence. Analysis of the effects of co-exposure to cadmium and PFOS confirmed the presence of an antagonistic effect. The inflection points of cadmium and PFOS were determined to be -1.11 and 2.27, respectively. Additionally, exposure to cadmium and PFOS had the opposite effects on two crucial pathways, namely, the rap1 and calcium signaling pathways, which involve core genes related to depression such as ADORA2A, FGF2, and FGFR1. These findings have significant implications for future studies and provide new strategies for exploring the mechanisms underlying co-exposure effects.


Alkanesulfonic Acids , Computational Biology , Depression , Environmental Pollutants , Fluorocarbons , Metals, Heavy , Fluorocarbons/toxicity , Metals, Heavy/toxicity , Humans , Alkanesulfonic Acids/toxicity , Environmental Pollutants/toxicity , Depression/epidemiology , Depression/chemically induced , Cadmium/toxicity , Nutrition Surveys , Environmental Exposure/adverse effects , Environmental Exposure/statistics & numerical data , Bayes Theorem , Decanoic Acids
9.
Mar Pollut Bull ; 203: 116446, 2024 Jun.
Article En | MEDLINE | ID: mdl-38703627

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.


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
10.
Cell Biochem Funct ; 42(4): e4060, 2024 Jun.
Article En | MEDLINE | ID: mdl-38816947

Perfluorooctane sulfonate (PFOS) is a pervasive organic toxicant that damages body organs, including heart. Isosakuranetin (ISN) is a plant-based flavonoid that exhibits a broad range of pharmacological potentials. The current investigation was conducted to evaluate the potential role of ISN to counteract PFOS-induced cardiac damage in rats. Twenty-four albino rats (Rattus norvegicus) were distributed into four groups, including control, PFOS (10 mg/kg) intoxicated, PFOS + ISN (10 mg/kg + 20 mg/kg) treated, and ISN (20 mg/kg) alone supplemented group. It was revealed that PFOS intoxication reduced the expressions of Nrf-2 and its antioxidant genes while escalating the expression of Keap-1. Furthermore, PFOS exposure reduced the activities of glutathione reductase (GSR), superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), glutathione S-transferase (GST), Heme oxygenase-1 (HO-1) and glutathione (GSH) contents while upregulating the levels of reactive oxygen species (ROS) and malondialdehyde (MDA). Besides, PFOS administration upregulated the levels of creatine kinase-MB (CK-MB), troponin I, creatine phosphokinase (CPK), and lactate dehydrogenase (LDH). Moreover, the levels of tumor necrosis factor-alpha (TNF-α), nuclear factor kappa-B (NF-κB), interleukin-6 (IL-6), and interleukin-1ß (IL-1ß) were increased after PFOS intoxication. Additionally, PFOS exposure downregulated the expression of Bcl-2 while upregulating the expressions of Bax and Caspase-3. Furthermore, PFOS administration disrupted the normal architecture of cardiac tissues. Nonetheless, ISN treatment remarkably protected the cardiac tissues via regulating aforementioned dysregulations owing to its antioxidative, anti-inflammatory, and antiapoptotic properties.


Alkanesulfonic Acids , Apoptosis , Fluorocarbons , Kelch-Like ECH-Associated Protein 1 , NF-E2-Related Factor 2 , Animals , Rats , Alkanesulfonic Acids/pharmacology , Alkanesulfonic Acids/toxicity , Apoptosis/drug effects , NF-E2-Related Factor 2/metabolism , Fluorocarbons/pharmacology , Kelch-Like ECH-Associated Protein 1/metabolism , Male , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/chemically induced , Inflammation/pathology , Flavones/pharmacology
11.
J Hazard Mater ; 470: 134177, 2024 May 15.
Article En | MEDLINE | ID: mdl-38565010

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.


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
12.
Environ Toxicol Pharmacol ; 107: 104434, 2024 Apr.
Article En | MEDLINE | ID: mdl-38582069

The potential toxic effects of linear alkylbenzene sulfonate (LAS), widely used in commercial detergents and cleaners, on submerged macrophytes remain unclear. We conducted a two-week exposure experiment to investigate LAS toxicity on five submerged macrophytes (four native and one exotic), focusing on their growth and physiological responses. The results showed that lower concentrations of LAS (< 5 mg/L) slightly stimulated the growth of submerged macrophytes, while higher doses inhibited it. Increasing LAS concentration resulted in decreased chlorophyll content, increased MDA content and POD activity, and initially increased SOD and CAT activities before declining. Moreover, Elodea nuttallii required a higher effective concentration for growth compared to native macrophytes. These findings suggest that different species of submerged macrophytes exhibited specific responses to LAS, with high doses (exceeding 5 ∼ 10 mg/L) inhibited plant growth and physiology. However, LAS may promote the dominance of surfactant-tolerant exotic submerged macrophytes in polluted aquatic environments.


Alkanesulfonic Acids , Antioxidants , Chlorophyll , Surface-Active Agents/toxicity , Alkanesulfonic Acids/toxicity
13.
Sci Total Environ ; 930: 172582, 2024 Jun 20.
Article En | MEDLINE | ID: mdl-38649052

Perfluoroalkyl sulfonate (PFOS) is a commonly used chemical compound that often found in materials such as waterproofing agents, food packaging, and fire retardants. Known for its stability and persistence in the environment, PFOS can enter the human body through various pathways, including water and the food chain, raising concerns about its potential harm to human health. Previous studies have suggested a cardiac toxicity of PFOS, but the specific cellular mechanisms remained unclear. Here, by using AC16 cardiomyocyte as a model to investigate the molecular mechanisms potential the cardiac toxicity of PFOS. Our findings revealed that PFOS exposure reduced cell viability and induces apoptosis in human cardiomyocyte. Proteomic analysis and molecular biological techniques showed that the Endoplasmic Reticulum (ER) stress-related pathways were activated, while the cellular autophagy flux was inhibited in PFOS-exposed cells. Subsequently, we employed strategies such as autophagy activation and ER stress inhibition to alleviate the PFOS-induced apoptosis in AC16 cells. These results collectively suggest that PFOS-induced ER stress activation and autophagy flux inhibition contribute to cardiomyocyte apoptosis, providing new insights into the mechanisms of PFOS-induced cardiomyocyte toxicity.


Alkanesulfonic Acids , Apoptosis , Autophagy , Endoplasmic Reticulum Stress , Fluorocarbons , Myocytes, Cardiac , Endoplasmic Reticulum Stress/drug effects , Myocytes, Cardiac/drug effects , Apoptosis/drug effects , Autophagy/drug effects , Fluorocarbons/toxicity , Alkanesulfonic Acids/toxicity , Humans , Cell Line , Environmental Pollutants/toxicity
14.
Ecotoxicol Environ Saf ; 276: 116318, 2024 May.
Article En | MEDLINE | ID: mdl-38626609

Perfluorooctane sulfonate (PFOS), an officially listed persistent organic pollutant, is a widely distributed perfluoroalkyl substance. Epidemiological studies have shown that PFOS is intimately linked to the occurrence of insulin resistance (IR). However, the detailed mechanism remains obscure. In previous studies, we found that mitochondrial calcium overload was concerned with hepatic IR induced by PFOS. In this study, we found that PFOS exposure noticeably raised lysosomal calcium in L-02 hepatocytes from 0.5 h. In the PFOS-cultured L-02 cells, inhibiting autophagy alleviated lysosomal calcium overload. Inhibition of mitochondrial calcium uptake aggravated the accumulation of lysosomal calcium, while inhibition of lysosomal calcium outflowing reversed PFOS-induced mitochondrial calcium overload and IR. Transient receptor potential mucolipin 1 (TRPML1), the calcium output channel of lysosomes, interacted with voltage-dependent anion channel 1 (VDAC1), the calcium intake channel of mitochondria, in the PFOS-cultured cells. Moreover, we found that ATP synthase F1 subunit beta (ATP5B) interacted with TRPML1 and VDAC1 in the L-02 cells and the liver of mice under PFOS exposure. Inhibiting ATP5B expression or restraining the ATP5B on the plasma membrane reduced the interplay between TRPML1 and VDAC1, reversed the mitochondrial calcium overload and deteriorated the lysosomal calcium accumulation in the PFOS-cultured cells. Our research unveils the molecular regulation of the calcium crosstalk between lysosomes and mitochondria, and explains PFOS-induced IR in the context of activated autophagy.


Alkanesulfonic Acids , Autophagy , Calcium , Fluorocarbons , Insulin Resistance , Liver , Lysosomes , Mitochondria , Mitochondrial Proton-Translocating ATPases , Alkanesulfonic Acids/toxicity , Fluorocarbons/toxicity , Animals , Lysosomes/drug effects , Lysosomes/metabolism , Autophagy/drug effects , Calcium/metabolism , Mice , Mitochondrial Proton-Translocating ATPases/metabolism , Liver/drug effects , Liver/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Male , Voltage-Dependent Anion Channel 1/metabolism , Cell Line , Hepatocytes/drug effects , Hepatocytes/metabolism , Environmental Pollutants/toxicity , TRPM Cation Channels/metabolism , Mice, Inbred C57BL
15.
Article En | MEDLINE | ID: mdl-38673379

Background: Exposure to environmental pollutants such as metals and Per- and Polyfluoroalkyl Substances (PFAS) has become common and increasingly associated with a decrease in the estimated Glomerular Filtration Rate (eGFR), which is a marker often used to measure chronic kidney disease (CKD). However, there are limited studies involving the use of both eGFR and the urine albumin creatinine ratio (uACR), which are more comprehensive markers to determine the presence of CKD and the complexity of pollutant exposures and response interactions, especially for combined metals and PFAS, which has not been comprehensively elucidated. Objective: This study aims to assess the individual and combined effects of perfluorooctanoic acid (PFOA), perfluorooctanesulfonic acid (PFOS), Cadmium (Cd), Mercury (Hg), and Lead (Pb) exposure on CKD using data from the National Health and Nutritional Examination Survey (NHANES) 2017-2018. Methods: We employed the use of bivariate logistic regression and Bayesian Kernel Machine Regression (BKMR) in our analysis of the data. Results: Logistic regression results revealed a positive association between PFOA and CKD. Our BKMR analysis revealed a non-linear and bi-phasic relationship between the metal exposures and CKD. In our univariate exposure-response function plot, Cd and Hg exhibited a U and N-shaped interaction, which indicated a non-linear and non-additive relationship with both low and high exposures associated with CKD. In addition, the bivariate exposure-response function between two exposures in a mixture revealed that Cd had a U-shaped relationship with CKD at different quantiles of Pb, Hg, PFOA, and PFOS, indicating that both low and high levels of Cd is associated with CKD, implying a non-linear and complex biological interaction. Hg's interaction plot demonstrated a N-shaped association across all quantiles of Cd, with the 75th quantile of Pb and the 50th and 75th quantiles of PFOA and PFOS. Furthermore, the PIP results underscored Cd's consistent association with CKD (PIP = 1.000) followed by Hg's (PIP = 0.9984), then PFOA and PFOS with a closely related PIP of 0.7880 and 0.7604, respectively, and finally Pb (PIP = 0.6940), contributing the least among the five environmental pollutants on CKD, though significant. Conclusions: Our findings revealed that exposure to environmental pollutants, particularly Hg and Cd, are associated with CKD. These findings highlight the need for public health interventions and strategies to mitigate the cumulative effect of PFAS and metal exposure and elucidate the significance of utilizing advanced statistical methods and tools to understand the impact of environmental pollutants on human health. Further research is needed to understand the mechanistic pathways of PFAS and metal-induced kidney injury and CKD, and longitudinal studies are required to ascertain the long-term impact of these environmental exposures.


Alkanesulfonic Acids , Cadmium , Caprylates , Environmental Exposure , Environmental Pollutants , Fluorocarbons , Lead , Renal Insufficiency, Chronic , Renal Insufficiency, Chronic/chemically induced , Renal Insufficiency, Chronic/epidemiology , Renal Insufficiency, Chronic/urine , Humans , Fluorocarbons/toxicity , Fluorocarbons/urine , Fluorocarbons/adverse effects , Environmental Pollutants/urine , Environmental Pollutants/toxicity , Female , Alkanesulfonic Acids/urine , Alkanesulfonic Acids/toxicity , Caprylates/toxicity , Caprylates/urine , Caprylates/adverse effects , Male , Cadmium/urine , Cadmium/toxicity , Middle Aged , Adult , Lead/urine , Lead/toxicity , Environmental Exposure/adverse effects , Nutrition Surveys , Mercury/urine , Mercury/toxicity , Aged , Bayes Theorem , Glomerular Filtration Rate/drug effects
16.
Toxicology ; 504: 153794, 2024 May.
Article En | MEDLINE | ID: mdl-38580097

Endocrine disruptors (EDs) pose a serious threat to human health and the environment and require a comprehensive evaluation to be identified. The identification of EDs require a substantial amount of data, both in vitro and in vivo, due to the current scientific criteria in the EU. At the same time, the EU strives to reduce animal testing due to concerns regarding animal welfare and sensitivity of animal studies to adequately detect adverse effects relevant for human health. Perfluorooctane sulfonic acid (PFOS) is a persistent organic pollutant that is suspected to be an ED based on academic research, however it is not identified as such from a regulatory perspective. It has previously been shown that PFOS has the potential to cause neurotoxicity as well as affect the thyroid system, and it is known that specific thyroid hormone levels are critical in the development of the brain during. In this work, the aim was to evaluate a mechanism-based approach to identify ED properties of PFOS based on the Adverse Outcome Pathway (AOP) framework and using New Approach Methods (NAMs), by comparing this approach to an ED assessment based on the currently available guidance document. An AOP network (AOPN) was generated for the thyroid modality, and AOPs leading to developmental neurotoxicity (DNT) were identified. A literature search and screening process based on the AOPN, and systematic review methodology, was performed, followed by a rigorous Weight-of-Evidence (WoE) assessment. Evidence was mapped back onto the AOPN used for the literature search, to identify possible endocrine Modes-of-Action (MoAs) for PFOS and data gaps in the two assessments. It could be concluded that PFOS fulfils the criteria for ED classification in the standard ED assessment, but not in the mechanism-based assessment. The need for quantitative information, such as quantitative AOPs, for the mechanism-based approach is discussed. The possibility of a directly neurotoxic alternative MoA was also highlighted based on available in vitro data. Opportunities and challenges with implementing AOPs and NAMs into the regulatory assessment of EDs, and assessing hazard in the Next Generation Risk Assessment, is discussed. This case study exploring the mechanism-based approach to ED identification represents an important step toward more accurate and predictive assessment of EDs based on AOPs and NAMs, and to the Next Generation Risk Assessment (NGRA) concept.


Adverse Outcome Pathways , Alkanesulfonic Acids , Endocrine Disruptors , Fluorocarbons , Animals , Humans , Alkanesulfonic Acids/toxicity , Endocrine Disruptors/toxicity , Environmental Pollutants/toxicity , Fluorocarbons/toxicity , Risk Assessment/methods
17.
Environ Int ; 187: 108687, 2024 May.
Article En | MEDLINE | ID: mdl-38677088

The biotoxicity of perfluoroalkyl and polyfluoroalkyl substances (PFASs) to aquatic organisms has been widely concerned. However, studies on toxic effects of PFASs are usually evaluated directly by using laboratory exposure rather than laboratory validation based on data obtained in the field. In this study, wild catfish (Silurus meridinalis) was explored on the relationship between PFASs bioaccumulation and lipid disorders. Nine and thirteen lipid metabolites were significantly associated with perfluorooctane sulfonate (PFOS) and 6:2/8:2Cl-PFESA (trade name F-53B) exposures, respectively; and the correlated lipid metabolites were the fatty acid (FA) and conjugates, FA esters, steroids, and glycerophosphate subclasses. The effects of PFASs on lipid metabolism of fish and its mechanism were further analyzed through exposure experiments. Zebrafish (Danio rerio) of different sexes underwent PFOS and F-53B exposures for 21 days at 100 ng/L and 100 µg/L. By determining gene expression levels, hepatic lipid contents, and histopathological change, the adverse effects order on lipid metabolism in male or female was 100 µg/L F-53B > 100 µg/L PFOS > 100 ng/L F-53B > 100 ng/L PFOS; the stress response in male was more intensive than that in female. PFOS and F-53B activated the peroxisome proliferator-activated receptor pathway, promoting the processes of FA and total cholesterol (T-CHO) transport, FA ß-oxidation, FA synthesis, and finally induced FA and T-CHO transportation from blood into liver, then accelerated FA to FA ester transformation, and CHO into steroids. Laboratory experiments confirmed the field analysis. This study innovatively explored the adverse effects of PFOS and F-53B on lipid metabolism and their mechanisms at field and laboratory levels, highlighting concerns regarding PFASs health risks.


Fluorocarbons , Lipid Metabolism , Water Pollutants, Chemical , Zebrafish , Animals , Lipid Metabolism/drug effects , Fluorocarbons/toxicity , Water Pollutants, Chemical/toxicity , Male , Female , Catfishes/metabolism , Alkanesulfonic Acids/toxicity , Liver/drug effects , Liver/metabolism
18.
Aquat Toxicol ; 271: 106923, 2024 Jun.
Article En | MEDLINE | ID: mdl-38669778

Perfluorooctanesulfonic acid (PFOS) is a ubiquitous legacy environmental contaminant detected broadly in human samples and water supplies. PFOS can cross the placenta and has been detected in cord blood and breastmilk samples, underscoring the importance of understanding the impacts of maternal PFOS exposure during early development. This study aimed to investigate the effects of a preconception exposure to PFOS on developmental endpoints in offspring, as well as examine the role of the transcription factor Nuclear factor erythroid-2-related factor (Nrf2a) in mediating these effects. This transcription factor regulates the expression of several genes that protect cells against oxidative stress including during embryonic development. Adult female zebrafish were exposed to 0.02, 0.08 or 0.14 mg/L PFOS for 1 week (duration of one cycle of oocyte maturation) and then paired with unexposed males from Nrf2a mutant or wildtype strains. Embryos were collected for two weeks or until completion of 5 breeding events. PFOS was maternally transferred to offspring independent of genotype throughout all breeding events in a dose-dependent manner, ranging from 2.77 to 23.72 ng/embryo in Nrf2a wildtype and 2.40 to 15.80 ng/embryo in Nrf2a mutants. Although embryo viability at collection was not impacted by maternal PFOS exposure, developmental effects related to nutrient uptake, growth and pancreatic ß-cell morphology were observed and differed based on genotype. Triglyceride levels were increased in Nrf2a wildtype eggs from the highest PFOS group. In Nrf2a wildtype larvae there was a decrease in yolk sac uptake while in Nrf2a mutants there was an increase. Additionally, there was a significant decrease in pancreatic ß-cell (islet) area in wildtype larvae from the 0.14 mg/L PFOS accompanied by an increase in the prevalence of abnormal islet morphologies compared to controls. Abnormal morphology was also observed in the 0.02 and 0.08 mg/L PFOS groups. Interestingly, in Nrf2a mutants there was a significant increase in the pancreatic ß-cell area in the 0.02 and 0.08 mg/L PFOS groups and no changes in the prevalence of abnormal islet morphologies. These results suggest that the regulation of processes like nutrient consumption, growth and pancreatic ß-cell development are at least partially modulated by the presence of a functional Nrf2a transcriptomic response. Overall, preconception exposure to environmental pollutants, such as PFOS, may impact the maturing oocyte and cause subtle changes that can ultimately impact offspring health and development.


Alkanesulfonic Acids , Fluorocarbons , Maternal Exposure , NF-E2-Related Factor 2 , Water Pollutants, Chemical , Zebrafish , Animals , Fluorocarbons/toxicity , Alkanesulfonic Acids/toxicity , Female , Water Pollutants, Chemical/toxicity , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Male , Embryo, Nonmammalian/drug effects , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Gene Expression Regulation, Developmental/drug effects , Embryonic Development/drug effects
19.
Ecotoxicol Environ Saf ; 277: 116368, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38669874

Perfluorooctane sulfonate (PFOS) is a persistent chemical that has long been a threat to human health. However, the molecular effects of PFOS on various organs are not well studied. In this study, male Sprague-Dawley rats were treated with various doses of PFOS through gavage for 21 days. Subsequently, the liver, lung, heart, kidney, pancreas, testis, and serum of the rats were harvested for lipid analysis. We applied a focusing lipidomic analytical strategy to identify key lipid responses of phosphorylcholine-containing lipids, including phosphatidylcholines and sphingomyelins. Partial least squares discriminant analysis revealed that the organs most influenced by PFOS exposure were the liver, kidney, and testis. Changes in the lipid profiles of the rats indicated that after exposure, levels of diacyl-phosphatidylcholines and 22:6-containing phosphatidylcholines in the liver, kidney, and testis of the rats decreased, whereas the level of 20:3-containing phosphatidylcholines increased. Furthermore, levels of polyunsaturated fatty acids-containing plasmenylcholines decreased. Changes in sphingomyelin levels indicated organ-dependent responses. Decreased levels of sphingomyelins in the liver, nonmonotonic dose responses in the kidney, and irregular responses in the testis after PFOS exposure are observed. These lipid responses may be associated with alterations pertaining to phosphatidylcholine synthesis, fatty acid metabolism, membrane properties, and oxidative stress in the liver, kidney, and testis. Lipid responses in the liver could have contributed to the observed increase in liver to body weight ratios. The findings suggest potential toxicity and possible mechanisms associated with PFOS in multiple organs.


Alkanesulfonic Acids , Fluorocarbons , Kidney , Liver , Rats, Sprague-Dawley , Testis , Animals , Alkanesulfonic Acids/toxicity , Fluorocarbons/toxicity , Male , Rats , Liver/drug effects , Liver/metabolism , Kidney/drug effects , Kidney/metabolism , Testis/drug effects , Testis/metabolism , Environmental Pollutants/toxicity , Sphingomyelins , Phosphatidylcholines , Lipid Metabolism/drug effects , Lipidomics , Lung/drug effects , Lung/metabolism
20.
Food Chem Toxicol ; 186: 114560, 2024 Apr.
Article En | MEDLINE | ID: mdl-38432440

Alpha lipoic acid (ALA) is a dietary supplement that has been used to treat a wide range of diseases, including obesity and diabetes, and have lipid-lowering effects, making it a potential candidate for mitigating dyslipidemia resulting from exposures to the per- and polyfluoroalkyl substance (PFAS) family member perfluorooctanesulfonic acid (PFOS). ALA can be considered a non-fluorinated structural analog to PFOS due to their similar 8-carbon chain and amphipathic structure, but, unlike PFOS, is rapidly metabolized. PFOS has been shown to reduce pancreatic islet area and induce ß-cell lipotoxicity, indicating that changes in ß-cell lipid microenvironment is a mechanism contributing to hypomorphic islets. Due to structural similarities, we hypothesized that ALA may compete with PFOS for binding to proteins and distribution throughout the body to mitigate the effects of PFOS exposure. However, ALA alone reduced islet area and fish length, with several morphological endpoints indicating additive toxicity in the co-exposures. Individually, ALA and PFOS increased fatty acid uptake from the yolk. ALA alone increased liver lipid accumulation, altered fatty acid profiling and modulated PPARÉ£ pathway signaling. Together, this work demonstrates that ALA and PFOS have similar effects on lipid uptake and metabolism during embryonic development in zebrafish.


Alkanesulfonic Acids , Fluorocarbons , Thioctic Acid , Water Pollutants, Chemical , Animals , Zebrafish , Thioctic Acid/pharmacology , Alkanesulfonic Acids/toxicity , Fluorocarbons/toxicity , Fatty Acids , Water Pollutants, Chemical/toxicity
...