Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.060
Filter
1.
Environ Health Perspect ; 132(9): 97004, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39258902

ABSTRACT

BACKGROUND: Although recent in vitro experimental results have raised the question of whether maternal exposure to per- and polyfluoroalkyl substances (PFAS) may be a potential environmental risk factor for chromosomal abnormalities, epidemiological studies investigating these associations are lacking. OBJECTIVES: This study examined whether prenatal PFAS exposure is associated with a higher prevalence of chromosomal abnormalities among offspring. METHODS: We used data from the Japan Environment and Children's Study, a nationwide birth cohort study, and employed logistic regression models to examine the associations between maternal plasma PFAS concentrations in the first trimester and the diagnosis of chromosomal abnormalities in all births (artificial abortions, miscarriages, stillbirths, and live births) up to 2 years of age. In addition, we examined associations with mixtures of PFAS using multipollutant models. RESULTS: The final sample consisted of 24,724 births with singleton pregnancies, of which 44 confirmed cases of chromosomal abnormalities were identified (prevalence: 17.8/10,000 births). When examined individually, exposure to perfluorononanoic acid (PFNA) and perfluorooctane sulfonic acid (PFOS) showed positive associations with any chromosomal abnormalities with age-adjusted odds ratios of 1.81 (95% CI: 1.26, 2.61) and 2.08 (95% CI: 1.41, 3.07) per doubling in concentration, respectively. These associations remained significant after Bonferroni correction, although they did not reach the adjusted significance threshold in certain sensitivity analyses. Furthermore, the doubling in all PFAS included as a mixture was associated with chromosomal abnormalities, indicating an age-adjusted odds ratio of 2.25 (95% CI: 1.34, 3.80), with PFOS as the predominant contributor, followed by PFNA, perfluoroundecanoic acid (PFUnA), and perfluorooctanoic acid (PFOA). DISCUSSION: The study findings suggested a potential association between maternal exposure to PFAS, particularly PFOS, and chromosomal abnormalities in offspring. However, the results should be interpreted cautiously, because selection bias arising from the recruitment of women in early pregnancy may explain the associations. https://doi.org/10.1289/EHP13617.


Subject(s)
Alkanesulfonic Acids , Chromosome Aberrations , Fluorocarbons , Maternal Exposure , Humans , Female , Japan/epidemiology , Fluorocarbons/blood , Fluorocarbons/toxicity , Pregnancy , Maternal Exposure/statistics & numerical data , Maternal Exposure/adverse effects , Chromosome Aberrations/chemically induced , Chromosome Aberrations/statistics & numerical data , Alkanesulfonic Acids/blood , Alkanesulfonic Acids/toxicity , Adult , Environmental Pollutants/toxicity , Environmental Pollutants/blood , Male , Infant , Infant, Newborn , Prenatal Exposure Delayed Effects/epidemiology , Prenatal Exposure Delayed Effects/chemically induced , Cohort Studies , Child, Preschool , Birth Cohort , Caprylates/toxicity , Caprylates/blood
2.
Food Funct ; 15(19): 10007-10019, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39282919

ABSTRACT

Background: Perfluorooctane sulfonate (PFOS) is a persistent, widely present environmental pollutant, and its toxicity to male reproduction has gradually attracted attention. Flaxseed oil (FO) is a dietary oil abundant in α-linolenic acid and has been demonstrated to possess multiple health benefits. However, whether FO protects against PFOS-induced testicular injury and its mechanism remain unclear. Methods: C57/BL6 mice were gavaged with different concentrations of FO or PFOS (10 mg kg-1) for 28 days. Blood and testicular tissues were collected for histopathology, proteomics, and biochemical and molecular analyses. Results: Our results showed that FO supplementation significantly attenuated PFOS-induced testicular injury, as indicated by histopathological changes, decreased oxidative stress level, increased sperm count, decreased rate of sperm malformation, and improved functional markers of spermatogenesis. Proteomic analysis showed that differentially expressed proteins were notably enriched in spliceosome pathways. Machine learning algorithms were used to screen the hub gene, and PRPF3 and PUF60 proteins were found to be important for FO to exert protective benefits to testicular injury. Western blot results confirmed that FO supplementation could increase the protein expression of PRPF3 and decrease the protein expression of PUF60 in PFOS-exposed mice. Conclusions: This study revealed that FO can alleviate PFOS-induced testicular dysfunction by regulating RNA alternative splicing. The spliceosome-related proteins PRPF3 and PUF60 may be the potential targets for FO to alleviate PFOS-induced testicular injury. FO supplementation may be an effective dietary intervention to prevent adverse effects of PFOS on testes.


Subject(s)
Alkanesulfonic Acids , Alternative Splicing , Fluorocarbons , Linseed Oil , Mice, Inbred C57BL , Testis , Male , Animals , Fluorocarbons/toxicity , Mice , Testis/drug effects , Testis/metabolism , Alkanesulfonic Acids/toxicity , Alternative Splicing/drug effects , Oxidative Stress/drug effects , Spermatozoa/drug effects , Spermatozoa/metabolism , Spermatogenesis/drug effects
3.
J Hazard Mater ; 479: 135676, 2024 Nov 05.
Article in English | MEDLINE | ID: mdl-39217921

ABSTRACT

Plants affect soil microorganisms through the release of root exudates under pollution stress. This process may affect rhizosphere priming effect (RPE) and alter the rate of soil organic matter decomposition. However, the influence of plants on the decomposition of organic matter in soil subjected to pollution stress remains unclear. We studied the effects of exposure to perfluorooctanesulfonic (PFOS) and its alternative, chlorinated polyfluoroalkyl ether sulfonic (F-53B), at concentrations of 0.1 mg/kg and 50 mg/kg on the RPE of reed. We conducted our experiments in an artificial climate chamber and used the natural 13C tracer method to determine RPE. In the PFOS-exposed groups, the RPE was negative, with values of -11.45 mg C kg-1 soil d-1 in the low PFOS group and -8.04 mg C kg-1 soil d-1 in the high PFOS group. In contrast, in the F-53B-exposed groups, the RPE was positive, with values of 8.26 mg C kg-1 soil d-1 in the low F-53B group and 12.18 mg C kg-1 soil d-1 in the high F-53B group. Exposure of reeds to PFOS/F-53B stress resulted in differential effects on extracellular enzyme activities. The observed positive and negative RPE phenomena could be attributed to variations in extracellular enzyme activities. In conclusion, RPE responded differently under PFOS/F-53B exposure.


Subject(s)
Alkanesulfonic Acids , Fluorocarbons , Rhizosphere , Soil Pollutants , Fluorocarbons/toxicity , Fluorocarbons/chemistry , Fluorocarbons/metabolism , Alkanesulfonic Acids/toxicity , Soil Pollutants/toxicity , Soil Pollutants/metabolism , Soil/chemistry , Poaceae/metabolism , Poaceae/drug effects , Soil Microbiology , Plant Roots/metabolism , Plant Roots/drug effects , Plant Roots/growth & development , Biodegradation, Environmental
4.
Chemosphere ; 365: 143374, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39306112

ABSTRACT

The widespread occurrence of perfluorooctane sulfonate (PFOS) and the mass production and application of graphene oxide (GO) lead to their inevitable release and interaction in the environment, which may enhance associated toxic impacts on aquatic organisms. This study elucidates the induction of apoptosis by 60-day chronic single and mixture exposures to environmentally relevant levels of PFOS (0.5 µg/L and 5 µg/L) and GO (1 mg/L) in adult marine medaka Oryzias melastigma. Results showed a significant increase (p < 0.05) in reactive oxygen species (ROS) levels, the apoptotic positive rate in livers, and activities of caspases 3, 8, and 9 in all treated groups compared to the control. PFOS individual and PFOS-GO combined exposures significantly impacted fish growth, upregulated expressions of six apoptosis-related genes including p53, apaf1, il1b, tnfa, bcl2l1, bax, as well as enriched cell cycle and p53 signaling pathways (transcriptomic analysis) related to apoptosis compared to control group. Besides higher ROS production, GO also had a higher binding affinity to proteins than PFOS, especially to caspase 8 as revealed by molecular docking. Overall, PFOS induced ROS-p53-caspase apoptosis pathway through multi-gene regulation during single or mixture exposure, while GO single exposure induced apoptosis through tissue damage and ROS-caspase pathway activation and direct docking with caspase 8 to activate the caspase cascade. Under co-exposure, the PFOS-induced apoptotic pathway overshadowed the GO-induced pathway, due to competition for limited active sites on caspases. These findings will contribute to a better understanding of the apoptosis mechanism and ecological risks of nanomaterials and per- and polyfluoroalkyl substances in marine ecosystems.


Subject(s)
Alkanesulfonic Acids , Apoptosis , Caspases , Fluorocarbons , Graphite , Oryzias , Reactive Oxygen Species , Tumor Suppressor Protein p53 , Water Pollutants, Chemical , Animals , Graphite/toxicity , Apoptosis/drug effects , Fluorocarbons/toxicity , Alkanesulfonic Acids/toxicity , Reactive Oxygen Species/metabolism , Water Pollutants, Chemical/toxicity , Oryzias/metabolism , Oryzias/physiology , Caspases/metabolism , Tumor Suppressor Protein p53/metabolism , Signal Transduction/drug effects , Molecular Docking Simulation , Environmental Exposure/adverse effects
5.
Chemosphere ; 365: 143396, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39313077

ABSTRACT

Per/polyfluoroalkyl substances (PFASs) are ubiquitous, bioaccumulative, and recalcitrant contaminants, posing global exposure and health risks. The effects of chemical structures on toxicities and the mechanisms of their obesogenic effects were largely unclear. This study used the model organism Caenorhabditis elegans to assess the impact of long-term exposure to different PFASs (PFNA, PFOSA, PFBS, PFHxS, 6:2 FTS, 4:2 FTS, PFOA, and PFOS) on growth and lipid metabolism and discussed the obesogenic mechanisms of selected PFASs. The growth assays indicated longer carbon-fluorine (-CF) chains and total fluorine atoms increased developmental toxicity of PFASs, while at 8 -CF chain-length, PFNA (-COOH terminal), PFOS (-SO3 terminal), and PFOSA (-SO2NH2 terminal) exhibited differential growth inhibition. With the toxicity ranking of PFNA > PFOS > PFOSA, all PFASs significantly induced total lipid accumulation and perturbed the lipid composition in C. elegans. All three PFASs significantly induced lipogenesis gene expression and partially suppressed lipolysis genes. The results suggested that the disruption of lipid metabolism of PFOSA depends on sbp-1, while PFNA and PFOS depend on nhr-49. In conclusion, long-term exposure to PFNA, PFOSA, and PFOS triggers obesogenic effects in organisms by distinct molecular mechanisms.


Subject(s)
Caenorhabditis elegans , Environmental Pollutants , Fluorocarbons , Lipid Metabolism , Animals , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/growth & development , Fluorocarbons/toxicity , Lipid Metabolism/drug effects , Environmental Pollutants/toxicity , Alkanesulfonic Acids/toxicity , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , Obesity , Lipogenesis/drug effects
6.
J Hazard Mater ; 478: 135515, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-39178777

ABSTRACT

The behavior and fate of PFOS (perfluorooctanesulfonate) in the aquatic environment have received great attention due to its high toxicity and persistence. The nanoscale supramolecular mechanisms of interaction between PFOS and ubiquitous EPS (exopolymers) remain unclear though EPS have been widely-known to influence the bioavailability of PFOS. Typically, the exposure patterns of PFOS in aquatic animals changed with the EPS-PFOS interaction are not fully understood. This study hypothesized that PFOS exposure and accumulation pathways depended on the PFOS-EPS interactive assembly behavior and animal species. Two model animals, zebrafish and chironomid larvae, with different feeding habitats were chosen for the exposure and accumulation tests at the environmental concentrations of PFOS in the absence and presence of EPS. It was found that PFOS triggered the self-assembly of EPS to form large aggregates which significantly trapped PFOS. PFOS accumulation was significantly promoted in zebrafish but drastically reduced in chironomid larvae because of the nanoscale interactive assembly between EPS and PFOS. The decreased dermal uptake but increased oral uptake of PFOS by zebrafish with large mouthpart size could be ascribed to the increased ingestion of PFOS-enriched EPS aggregates as food. For the chironomid larvae with small mouthpart size, the PFOS-EPS assemblies reduced the dermal, oral and intestinal uptake of PFOS. The nano-visualization evidences confirmed that the PFOS-enriched EPS-PFOS assemblies blocked PFOS penetration through skin of both animals. These findings provide novel knowledge about the ecological risk of PFOS in aquatic environments.


Subject(s)
Alkanesulfonic Acids , Chironomidae , Fluorocarbons , Larva , Water Pollutants, Chemical , Zebrafish , Animals , Alkanesulfonic Acids/metabolism , Alkanesulfonic Acids/toxicity , Fluorocarbons/metabolism , Fluorocarbons/toxicity , Chironomidae/metabolism , Chironomidae/drug effects , Water Pollutants, Chemical/toxicity , Water Pollutants, Chemical/metabolism , Larva/metabolism , Larva/drug effects , Ecosystem
7.
J Hazard Mater ; 478: 135583, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-39180998

ABSTRACT

Perfluorooctanoate (PFOA) and perfluorooctane sulfonate (PFOS), two prominent per- and polyfluoroalkyl substances (PFASs), are potentially harmful to many human organs. However, there only exist limited methods to mitigate their health hazards. The aim of this study is to combine a bioinformatics analysis with in vitro experiments to discover small molecules that can alleviate liver damage caused by PFOA/PFOS. We identified 192 and 82 key genes related to hepatocytes exposed to PFOA and PFOS, respectively. The functional enrichment analysis of key genes suggested cellular senescence may be important in PFOA/PFOS-induced hepatotoxicity. The in vitro models revealed that PFOA/PFOS led to hepatocyte senescence by increasing the activity of SA-ß-gal, inducing mitochondrial dysfunction, impacting cell cycle arrest, and elevating the expressions of p21, p53, IL-1ß, and SASP-related cytokines. The drug-target gene set enrichment analysis method was employed to compare the transcriptome data from the Gene Expression Omnibus database (GEO), Comparative Toxicogenomics Database (CTD), and the high-throughput experiment- and reference-guided database (HERB), and 21 traditional Chinese medicines (TCMs) were identified that may alleviate PFOA/PFOS-induced liver aging. The experimental results of co-exposure to PFOA/PFOS and TCMs showed that sanguinarine has particular promise in alleviating cellular senescence caused by PFOA/PFOS. Further investigations revealed that the mTOR-p53 signaling pathway was involved in PFOA/PFOS-mediated hepatic senescence and can be blocked using sanguinarine.


Subject(s)
Alkanesulfonic Acids , Caprylates , Cellular Senescence , Fluorocarbons , Hepatocytes , Isoquinolines , Fluorocarbons/toxicity , Hepatocytes/drug effects , Hepatocytes/metabolism , Cellular Senescence/drug effects , Caprylates/toxicity , Humans , Alkanesulfonic Acids/toxicity , Isoquinolines/pharmacology , Benzophenanthridines/pharmacology , Computational Biology , Animals , Hep G2 Cells , Signal Transduction/drug effects
10.
Sci Total Environ ; 951: 175716, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-39181253

ABSTRACT

Perfluorooctane sulfonate (PFOS) and its precursor, perfluorooctane sulfonamide (PFOSA), are widespread in the environment. Evidence suggests a strong link between maternal exposure to PFOS/PFOSA and congenital heart diseases in the offspring, but the underlying mechanisms remain unclear. We hypothesized that PFOS and PFOSA induce cardiac defects through the peroxisome proliferator-activated receptor gamma (PPARγ) and aryl hydrocarbon receptor (AHR) pathways, respectively. In this study, we demonstrated that exposing zebrafish embryos to either PFOSA or PFOS caused cardiac malformations and dysfunction. Both PFOS and PFOSA induced reactive oxygen species (ROS) overproduction, mitochondrial damage, and apoptosis in zebrafish larvae hearts. Blockade of PPARγ through either pharmaceutical inhibition or genetic knockdown only attenuated the changes caused by PFOS, but not those elicited by PFOSA. Conversely, inhibition of AHR alleviated the adverse effects induced by PFOSA but not by PFOS. Both PFOSA and PFOS exhibited similar binding affinities to AHR using molecular docking techniques. The varying ability of PFOS and PFOSA to induce AHR activity in zebrafish embryonic hearts can be attributed to their different capabilities for activating PPARγ. In summary, our findings indicate that PFOS and PFOSA induce excessive ROS production in zebrafish larvae via the PPARγ and AHR pathways, respectively. This oxidative stress in turn causes mitochondrial damage and apoptosis, leading to cardiac defects.


Subject(s)
Alkanesulfonic Acids , Fluorocarbons , Oxidative Stress , PPAR gamma , Receptors, Aryl Hydrocarbon , Zebrafish , Animals , Fluorocarbons/toxicity , Receptors, Aryl Hydrocarbon/metabolism , Oxidative Stress/drug effects , Alkanesulfonic Acids/toxicity , PPAR gamma/metabolism , Water Pollutants, Chemical/toxicity , Heart Defects, Congenital/chemically induced , Reactive Oxygen Species/metabolism , Embryo, Nonmammalian/drug effects , Sulfonamides/toxicity
11.
Sci Total Environ ; 951: 175612, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-39163934

ABSTRACT

Perfluorooctane sulfonate (PFOS) is a persistent organic pollutant widely utilized in industrial manufacturing and daily life, leading to significant environmental accumulation and various public health issues. This study aims to characterize spliceosome-associated protein 130 (SAP130) as a key mediator of crosstalk between hepatocytes and macrophages, elucidating its role in PFOS-induced liver inflammation. The data demonstrate that PFOS exposure induces ferroptosis in mouse liver and AML12 cells. During ferroptosis, SAP130 is released from injured hepatocytes into the microenvironment, binding to macrophage-inducible C-type lectin (Mincle) and activating the Mincle/Syk signaling pathway in macrophages, ultimately promoting M1 polarization and exacerbating liver injury. Treatment with the ferroptosis inhibitor Ferrostatin-1 reduces SAP130 release, inhibits Mincle/Syk signaling activation, and mitigates inflammatory response. Furthermore, siSAP130 suppresses the activation of the Mincle signaling pathway and M1 polarization in BMDM cells. Conversely, treatment with the ferroptosis agonist Erastin enhances paracrine secretion of SAP130 and exacerbates inflammation. These findings emphasize the significance of hepatocyte-macrophage crosstalk as a critical pathway for PFOS-induced liver injury in mice while highlighting SAP130 as a pivotal regulator of ferroptosis and inflammation, thereby elucidating the potential mechanism of PFOS-induced liver injury.


Subject(s)
Alkanesulfonic Acids , Ferroptosis , Fluorocarbons , Hepatocytes , Macrophages , Ferroptosis/drug effects , Ferroptosis/physiology , Animals , Fluorocarbons/toxicity , Mice , Hepatocytes/drug effects , Macrophages/drug effects , Alkanesulfonic Acids/toxicity , Chemical and Drug Induced Liver Injury , Environmental Pollutants/toxicity , Signal Transduction/drug effects
12.
J Hazard Mater ; 477: 135370, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39088956

ABSTRACT

Linear alkylbenzene sulfonate (LAS) is one of the most widely used anionic surfactants and a common toxic pollutant in wastewater. This study employed high throughput sequencing to explore the microbial community structure within activated sludge exposed to a high concentration of LAS. Genera such as Pseudomonas, Aeromonas, Thauera and Klebsiella exhibited a significant positive correlation with LAS concentrations. Furthermore, Comamonas and Klebsiella were significantly enriched under the stress of LAS. Moreover, bacterial strains with LAS-degrading capability were isolated and characterized to elucidate the degradation pathways. The Klebsiella pneumoniae isolate L1 could effectively transform more than 60 % of 25 mg/L of LAS within 72 h. Chemical analyses revealed that L1 utilized the LAS sulfonyl group as a sulfur source to support its growth. Genomic and transcriptomic analyses suggested that strain L1 may uptake LAS through the sulfate ABC transport system and remove sulfonate with sulfate and sulfite reductases.


Subject(s)
Alkanesulfonic Acids , Biodegradation, Environmental , Sewage , Surface-Active Agents , Surface-Active Agents/metabolism , Surface-Active Agents/toxicity , Alkanesulfonic Acids/metabolism , Alkanesulfonic Acids/toxicity , Sewage/microbiology , Bacteria/metabolism , Bacteria/genetics , Bacteria/drug effects , Water Pollutants, Chemical/metabolism , Water Pollutants, Chemical/toxicity , Microbiota/drug effects
13.
Ecotoxicol Environ Saf ; 284: 116924, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39181077

ABSTRACT

Perfluorooctane sulfonate (PFOS) is a persistent organic pollutant and accumulated in the liver of mammals. PFOS exposure is closely associated with the development of pyroptosis. Nevertheless, the underlying mechanism is unclear. We found here that PFOS induced pyroptosis in the mice liver and L-02 cells as demonstrated by activation of the NOD-like receptor protein 3 inflammasome, gasdermin D cleavage and increased release of interleukin-1ß and interleukin-18. The level of cytoplasmic calcium was accelerated in hepatocytes upon exposure to PFOS. The phosphorylated/activated form of calcium/calmodulin-dependent protein kinase II (CaMKII) was augmented by PFOS in vivo and in vitro. PFOS-induced pyroptosis was relieved by CaMKII inhibitor. Among various CaMKII subtypes, we identified that CaMKIIγ was activated specifically by PFOS. CaMKIIγ interacted with Smad family member 3 (Smad3) under PFOS exposure. PFOS increased the phosphorylation of Smad3, and CaMKII inhibitor or CaMKIIγ siRNA alleviated PFOS-caused phosphorylation of Smad3. Inhibiting Smad3 activity was found to alleviate PFOS-induced hepatocyte pyroptosis. This study puts forward that CaMKIIγ-Smad3 is the linkage between calcium homeostasis disturbance and pyroptosis, providing a mechanistic explanation for PFOS-induced pyroptosis.


Subject(s)
Alkanesulfonic Acids , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Fluorocarbons , Hepatocytes , Pyroptosis , Smad3 Protein , Alkanesulfonic Acids/toxicity , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Fluorocarbons/toxicity , Phosphorylation , Smad3 Protein/metabolism , Mice , Pyroptosis/drug effects , Mice, Inbred C57BL , Male , Environmental Pollutants/toxicity
14.
Ecotoxicol Environ Saf ; 284: 116890, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39146593

ABSTRACT

Perfluorooctane sulfonate (PFOS) is known as a persistent organic pollutant. A significant correlation between PFOS and liver ferroptosis has been unveiled, but the precise mechanism needs to be elucidated. In prior research, we found that PFOS treatment provoked mitochondrial iron overload. In this study, we observed a gradual increase in lysosomal iron in L-O2 cells after exposure to PFOS for 0.5-24 h. In PFOS-exposed L-O2 cells, suppressing autophagy relieved the lysosomal iron overload. Inhibiting transient receptor potential mucolipin 1 (TRPML1), a calcium efflux channel on the lysosomal membrane, led to a further rise in lysosomal iron levels and decreased mitochondrial iron overload during PFOS treatment. Suppressing VDAC1, a subtype of voltage-dependent anion-selective channels (VDACs) on the outer mitochondrial membrane, had no impact on PFOS-triggered mitochondrial iron overload, whereas restraining VDAC2/3 relieved this condition. Although silencing VDAC2 relieved PFOS-induced mitochondrial iron overload, it had no effect on PFOS-triggered lysosomal iron overload. Silencing VDAC3 alleviated PFOS-mediated mitochondrial iron overload and led to an additional increase in lysosomal iron. Therefore, we regarded VDAC3 as the specific VDACs subtype that mediated the lysosomes-mitochondria iron transfer. Additionally, in the presence of PFOS, an enhanced association between TRPML1 and VDAC3 was found in mice liver tissue and L-O2 cells. Our research unveils a novel regulatory mechanism of autophagy on the iron homeostasis and the effect of TRPML1-VDAC3 interaction on lysosomes-mitochondria iron transfer, giving an explanation of PFOS-induced ferroptosis and shedding some light on the role of classic calcium channels in iron transmission.


Subject(s)
Alkanesulfonic Acids , Ferroptosis , Fluorocarbons , Hepatocytes , Iron , Lysosomes , Mitochondria , Ferroptosis/drug effects , Lysosomes/drug effects , Lysosomes/metabolism , Alkanesulfonic Acids/toxicity , Fluorocarbons/toxicity , Animals , Iron/metabolism , Mice , Hepatocytes/drug effects , Hepatocytes/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Cell Line , Environmental Pollutants/toxicity , Mice, Inbred C57BL , Male , Autophagy/drug effects , Iron Overload
15.
Environ Health Perspect ; 132(8): 87007, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39177951

ABSTRACT

BACKGROUND: Previous epidemiological studies have repeatedly found per- and polyfluoroalkyl substances (PFAS) exposure associated with higher circulating cholesterol, one of the greatest risk factors for development of coronary artery disease. The main route of cholesterol catabolism is through its conversion to bile acids, which circulate between the liver and ileum via enterohepatic circulation. Patients with coronary artery disease have decreased bile acid excretion, indicating that PFAS-induced impacts on enterohepatic circulation may play a critical role in cardiovascular risk. OBJECTIVES: Using a mouse model with high levels of low-density and very low-density lipoprotein (LDL and VLDL, respectively) cholesterol and aortic lesion development similar to humans, the present study investigated mechanisms linking exposure to a PFAS mixture with increased cholesterol. METHODS: Male and female Ldlr-/- mice were fed an atherogenic diet (Clinton/Cybulsky low fat, 0.15% cholesterol) and exposed to a mixture of 5 PFAS representing legacy, replacement, and emerging subtypes (i.e., PFOA, PFOS, PFHxS, PFNA, GenX), each at a concentration of 2mg/L, for 7 wk. Blood was collected longitudinally for cholesterol measurements, and mass spectrometry was used to measure circulating and fecal bile acids. Transcriptomic analysis of ileal samples was performed via RNA sequencing. RESULTS: After 7 wk of PFAS exposure, average circulating PFAS levels were measured at 21.6, 20.1, 31.2, 23.5, and 1.5µg/mL in PFAS-exposed females and 12.9, 9.7, 23, 14.3, and 1.7µg/mL in PFAS-exposed males for PFOA, PFOS, PFHxS, PFNA, and GenX, respectively. Total circulating cholesterol levels were higher in PFAS-exposed mice after 7 wk (352mg/dL vs. 415mg/dL in female mice and 392mg/dL vs. 488mg/dL in male mice exposed to vehicle or PFAS, respectively). Total circulating bile acid levels were higher in PFAS-exposed mice (2,978 pg/µL vs. 8,496 pg/µL in female mice and 1,960 pg/µL vs. 4,452 pg/µL in male mice exposed to vehicle or PFAS, respectively). In addition, total fecal bile acid levels were lower in PFAS-exposed mice (1,797 ng/mg vs. 682 ng/mg in females and 1,622 ng/mg vs. 670 ng/mg in males exposed to vehicle or PFAS, respectively). In the ileum, expression levels of the apical sodium-dependent bile acid transporter (ASBT) were higher in PFAS-exposed mice. DISCUSSION: Mice exposed to a PFAS mixture displayed higher circulating cholesterol and bile acids perhaps due to impacts on enterohepatic circulation. This study implicates PFAS-mediated effects at the site of the ileum as a possible critical mediator of increased cardiovascular risk following PFAS exposure. https://doi.org/10.1289/EHP14339.


Subject(s)
Bile Acids and Salts , Fluorocarbons , Animals , Bile Acids and Salts/metabolism , Mice , Fluorocarbons/toxicity , Male , Female , Receptors, LDL/genetics , Receptors, LDL/metabolism , Environmental Pollutants/toxicity , Lipids/blood , Cholesterol/blood , Cholesterol/metabolism , Alkanesulfonic Acids/toxicity
16.
Chemosphere ; 364: 143075, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39151576

ABSTRACT

Recently, PFASs toxicity for the human immune system has become a growing concern. However, there is currently limited information on PFASs immunotoxicity beyond PFHxS, PFOA, PFOS, and PFNA. Therefore, it is urgent to close the present knowledge gap by testing a wider range of compounds. In the present study, twelve compounds were tested for a relationship between the chain-length and headgroup of a PFAS and its cytotoxic for THP-1. As such, THP-1, either as monocytes or differentiated macrophages, were exposed to PFASs in a concentration range of 0-800 µM for either 3 or 24 h. After that, cell viability and reactive oxygen species (ROS) generation were assessed using MTT and DCFH assay, respectively. PFASs' cytotoxicity is dependent on both their chain-length and headgroups. Cell viability decreased with increasing chain-length, and FTOHs displayed markedly higher toxicity than PFCAs and PFSAs. PFASs were ranked based on their calculated Relative Potency Factor. The ranking for the cytotoxicity data on monocytes appears to be 6:2 FTOH â‰« PFNA > PFDA > PFOS > PFOA >4: 2 FTOH > PFHxS = PFHxA > PFBA. For macrophages, this ranking was as follows: 6:2 FTOH >4:2 FTOH > PFOS > PFDA > PFNA > PFOA > PFHxS. The results observed for the ROS generating potential differed as FTOHs generated no ROS. Here, the ranking in monocytes was PFOA > PFNA > PFOS > PFHxS > PFDA > PFHxA = PFBS = PFBA. The ranking for macrophages was PFNA > PFDA ≥ PFOA > PFOS > PFHxA > PFHxS > PFBA = PFBS. In conclusion, the carbon chain-length and functional headgroup of a PFAS are major determinants for their toxicity to THP-1 cells. Furthermore, our study demonstrates the most potent cytotoxic effect for FTOHs in vitro, which has not been observed before to the authors' knowledge.


Subject(s)
Cell Survival , Fluorocarbons , Macrophages , Monocytes , Reactive Oxygen Species , Fluorocarbons/toxicity , Fluorocarbons/chemistry , Humans , Macrophages/drug effects , Monocytes/drug effects , Reactive Oxygen Species/metabolism , Cell Survival/drug effects , Structure-Activity Relationship , THP-1 Cells , Alkanesulfonic Acids/toxicity , Alkanesulfonic Acids/chemistry , Environmental Pollutants/toxicity , Environmental Pollutants/chemistry
17.
Environ Int ; 191: 108962, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39159514

ABSTRACT

Exposed to ubiquitously perfluorooctanoic acid (PFOA) and perfluorooctane sulfonate (PFOS) has been associated with non-alcoholic fatty liver disease (NAFLD), yet the underlying molecular mechanism remains elusive. The extrapolation of empirical studies correlating per- and polyfluoroalkyl substance (PFAS) exposure with NAFLD occurrence to real-life exposure was hindered by the limited availability of mechanistic data at environmentally relevant concentrations. Herein, a novel pathway mediating hepatocyte lipid accumulation by PFOA and PFOS at human-relevant dose (<10 µM) was identified by integrating CRISPR-Cas9 genome screening, concentration-dependent transcriptional assay in HepG2 cell and epidemiological data mining. 1) At genetic level, nudt7 showed the highest enriched potency among 569 NAFLD-related genes, and the transcription of nudt7 was significantly downregulated by PFOA and PFOS exposure (<7 µM). 2) At molecular pathway, upon exposure to ≤10-4 µM PFOA and PFOS, the downregulation of nudt7 transcriptional expression triggered the reduction of Ace-CoA hydrolase activity. 3) At cellular level, increased lipids were measured in HepG2 cells with PFOA and PFOS (<2 µM). Overall, we identified a novel mechanism mediated by transcriptional downregulation of nudt7 gene in hepatocellular lipid increase treated with PFOA and PFOS, which could potentially explain the NAFLD occurrence associated with exposure to PFASs in humans.


Subject(s)
Alkanesulfonic Acids , Caprylates , Fluorocarbons , Hepatocytes , Lipid Metabolism , Humans , Fluorocarbons/toxicity , Alkanesulfonic Acids/toxicity , Caprylates/toxicity , Hepatocytes/drug effects , Hepatocytes/metabolism , Hep G2 Cells , Lipid Metabolism/drug effects , Environmental Pollutants/toxicity , Non-alcoholic Fatty Liver Disease/chemically induced , Non-alcoholic Fatty Liver Disease/metabolism
18.
Sci Total Environ ; 952: 175726, 2024 Nov 20.
Article in English | MEDLINE | ID: mdl-39181257

ABSTRACT

Per- and poly-fluoroalkyl substances (PFAS) are an emerging class of persistent organic pollutants that are widespread in aquatic ecosystems and pose a serious threat to aquatic organisms. It is thus crucial to explore the toxicity mechanisms of PFAS to submerged macrophytes and biofilms. In this study, Vallisneria natans (V. natans) was exposed to environmentally relevant concentrations of perfluorooctanoic acid (PFOA) and perfluorooctane sulphonate (PFOS). Results showed that PFAS induced the excessive production of reactive oxygen species, triggering antioxidant responses. V. natans exhibited an improved stress tolerance by altering the biosynthesis of several plant secondary metabolites and the histidine, arginine, proline pathways in response to PFAS exposure. Moreover, PIP1-1, PIP2-2, SLAH1 and SLAH2 genes were upregulated, indicating the activation of aquaporins and slow-type anion channels. The uptake of PFOA and PFOS by V. natans was 41.74 % and 52.31 %, respectively. Notably, PFAS bound to functional proteins (GSTF10), promoting the detoxification of plants. Exposure to PFAS also altered the structure of biofilms by inducing the synthesis of large amounts of polysaccharides and proteins. The diversity and richness of the microbial community within periphytic biofilms changed significantly. These results provide a comprehensive description of the responses of aquatic plants and periphytic biofilms to PFAS and the removal mechanism of PFAS, contributing to the environmental risk assessments and removal of PFAS in aquatic ecosystems.


Subject(s)
Biofilms , Fluorocarbons , Water Pollutants, Chemical , Biofilms/drug effects , Water Pollutants, Chemical/toxicity , Fluorocarbons/toxicity , Alkanesulfonic Acids/toxicity , Caprylates/toxicity , Hydrocharitaceae/metabolism , Hydrocharitaceae/drug effects
19.
Toxicol Ind Health ; 40(12): 653-666, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39217409

ABSTRACT

Perfluorooctane sulfonate (PFOS) is one of the most widely used perfluorinated compounds, and as an environmental endocrine disruptor and environmental persistent pollutant, the threat of PFOS to human health is of increasing concern. Exposure to PFOS has been shown to be closely associated with liver disease, but the intrinsic molecular targets and mechanisms of PFOS-induced liver damage are not well understood. This study was conducted to explore whether the Wnt/ß-Catenin signaling pathway and the endoplasmic reticulum stress signaling pathway are involved in damage of PFOS to the liver. In this study, we used the CCK-8 method to detect cell viability, a microscope and DAPI staining to observe cell morphology, flow cytometry to detect cell ROS and apoptosis levels; and Western blot to detect the expressions of proteins in the WNT/ß-Catenin, endoplasmic reticulum stress and apoptosis-related pathways. We found that PFOS activated WNT/ß-Catenin and endoplasmic reticulum stress-related pathways in L-02 cells and could lead to the development of oxidative stress and apoptosis. Our findings showed that PFOS could cause damage to L-02 cells, and the WNT/ß-Catenin signaling and endoplasmic reticulum stress pathways were involved in the changes caused by PFOS to L-02 cells, which provided a new theoretical basis for studying the hepatotoxicity and mechanism of PFOS. PFOS can lead to increased intracellular ROS levels, causing oxidative stress, endoplasmic reticulum stress and activation of the WNT/ß-catenin signaling pathway. Our experimental results showed that PFOS can cause damage to L-02 cells, and the WNT/ß-Catenin signaling pathway and endoplasmic reticulum stress pathway are involved in the process of damage caused by PFOS to L-02 cells.


Subject(s)
Alkanesulfonic Acids , Apoptosis , Endoplasmic Reticulum Stress , Fluorocarbons , Wnt Signaling Pathway , Alkanesulfonic Acids/toxicity , Fluorocarbons/toxicity , Endoplasmic Reticulum Stress/drug effects , Humans , Wnt Signaling Pathway/drug effects , Cell Line , Apoptosis/drug effects , Cell Survival/drug effects , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , beta Catenin/metabolism , Environmental Pollutants/toxicity
20.
Article in English | MEDLINE | ID: mdl-39054005

ABSTRACT

Per- and polyfluoroalkyl substances (PFAS) comprise many chemicals with strong carbon-carbon and carbon-fluorine bonds and have extensive industrial applications in manufacturing several consumer products. The solid covalent bonding makes them more persistent in the environment and stays away from all types of degradation, naming them 'forever chemicals.' Zebrafish (Danio rerio) was used to evaluate the genotoxic and cytotoxic effects of legacy PFAS, Perfluorooctane sulfonate (PFOS), and its alternatives, such as Perfluoro-2-methyl-3-oxahexanoic acid ammonium (GenX) and 7H-Perfluoro-3,6-dioxa-4-methyl-octane-1-sulfonic acid (Nafion by-product 2 [NBP2]) upon single and combined exposure at an environmental concentration of 10 µg/L for 48-h. Erythrocyte micronucleus cytome assay (EMNCA) revealed an increased frequency of micronuclei (MN) in fish erythrocytes with a significant increase in NBP2-treated fish. The order of genotoxicity noticed was NBP2 > PFOS > Mixture > GenX in D. rerio. Fish exposed to PFOS and its alternatives in single and combined experiments did not cause any significant difference in nuclear abnormalities. However, PFOS and combined exposure positively inhibit cytokinesis, resulting in an 8.16 and 7.44-fold-change increase of binucleated cells. Besides, statistically, increased levels of reactive oxygen species (ROS) and malondialdehyde (MDA) content indicate oxidative stress in D. rerio. In addition, 'forever chemicals' resulted in cytotoxicity, as evident through changes in nucleus width to the erythrocyte length in NBP2 and mixture exposure groups. The findings revealed that PFAS alternative NBP2 is more toxic than PFOS in inducing DNA damage and cytotoxicity. In addition, all three tested 'forever chemicals' induced ROS and lipid peroxidation after individual and combined exposure. The present work is the first to concern the genotoxicity and cytotoxicity of 'forever chemicals' in the aquatic vertebrate D. rerio.


Subject(s)
Alkanesulfonic Acids , DNA Damage , Fluorocarbons , Micronucleus Tests , Water Pollutants, Chemical , Zebrafish , Animals , Fluorocarbons/toxicity , Micronucleus Tests/methods , Alkanesulfonic Acids/toxicity , DNA Damage/drug effects , Water Pollutants, Chemical/toxicity , Oxidative Stress/drug effects , Erythrocytes/drug effects , Reactive Oxygen Species/metabolism , Mutagens/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL