Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 159
Filter
1.
Cell Biochem Funct ; 42(4): e4060, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38816947

ABSTRACT

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.


Subject(s)
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
2.
Bioresour Technol ; 393: 129999, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37980946

ABSTRACT

Per- and polyfluoroalkyl substances (PFASs) are recalcitrant organic pollutants, which accumulate widely in aquatic and solid matrices. Anaerobic digestion (AD) is one of possible options to manage organic wastes containing PFASs, however, the impacts of different types of PFAS on AD remains unclear. This study aimed to critically investigate the effects of two representative PFAS compounds, i.e., perfluorooctanoic acid (PFOA) and perfluorooctane sulfonate (PFOS), on the AD performance and microbial community structure. 100 mg/L of both PFOA and PFOS considerably inhibited the AD performance and changed the microbial community structure. Especially, PFOA was more toxic to bacterial and archaeal activity than PFOS, which was reflected in AD performance. In addition, the sulfonic acid group in PFOS affected the changes in microbial community structure by inducing abundant sulfate reducing bacteria (i.e., Desulfobacterota). This study provides a significant reference to the response of AD system on different PFAS types and dosage.


Subject(s)
Alkanesulfonic Acids , Caprylates , Environmental Pollutants , Fluorocarbons , Microbiota , Anaerobiosis , Alkanesulfonic Acids/pharmacology , Fluorocarbons/toxicity , Fluorocarbons/analysis
3.
Environ Sci Technol ; 57(22): 8213-8224, 2023 06 06.
Article in English | MEDLINE | ID: mdl-37216669

ABSTRACT

Epidemiological evidence regarding the effects of prenatal exposure to perfluoroalkyl substances (PFASs) on neurodevelopment in children is inconclusive. In 449 mother-child pairs from the Shanghai-Minhang Birth Cohort Study, we measured the concentrations of 11 PFASs in maternal plasma samples obtained at 12-16 weeks of gestation. We assessed children's neurodevelopment at 6 years of age by the fourth edition of the Chinese Wechsler Intelligence Scale for Children and Child Behavior Checklist for ages 6-18. We evaluated the association between prenatal exposure to PFASs and children's neurodevelopment and the effect modification of maternal dietary factors during pregnancy and the child's sex. We found that prenatal exposure to multiple PFASs was associated with increased scores for attention problems, and the individual effect of perfluorooctanoic acid (PFOA) was statistically significant. However, no statistically significant association between PFASs and cognitive development was observed. Additionally, we found the effect modification of maternal nut intake and child's sex. In conclusion, this study suggests that prenatal exposure to PFASs was associated with more attention problems, and maternal nut intake during pregnancy may alter the potential effect of PFASs. However, these findings were exploratory because of multiple testing and the relatively small sample size.


Subject(s)
Alkanesulfonic Acids , Environmental Pollutants , Fluorocarbons , Prenatal Exposure Delayed Effects , Female , Pregnancy , Humans , Prenatal Exposure Delayed Effects/epidemiology , Cohort Studies , China , Cognition , Alkanesulfonic Acids/pharmacology , Maternal Exposure
4.
Immunobiology ; 228(3): 152356, 2023 05.
Article in English | MEDLINE | ID: mdl-36857909

ABSTRACT

Perfluorooctane sulfonic acid (PFOS) is member of a class of molecules with fluorinated carbon chains known as polyfluoroalkyls. PFOS have been used to produce a variety of industry and comsumer uses. However, a significant concern is that it accumulates in the environment, including in animals and humans, and that it is a potential immunosuppressant. Here we analyze immune homeostasis in mice following chronic exposure to PFOS at levels up to those historically found in PFOS manufacturing workers. Mice were exposed to 0.15, 1.5, 15, or 50 µg /kg of PFOS for 28 days, after which, B cells, T cells, and granulocytes from the bone marrow, liver, spleen, lymph nodes, and thymus were evaluated. We find that at these exposures, there was no effect of PFOS on major T- or B-cell populations, macrophages, dendritic cells, basophils, mast cells, eosinophils, neutrophils, serum antibodies or select serum cytokines. By contrast, mice exposed the known immunosuppressant cyclophosphamide, which was given at 40 mg/kg for four days, exhibited depletion of several granulocyte, T- and B-cell populations of the thymus, bone marrow, and spleen, as well as circulating IgM and IgE antibodies. These data indicate that exposures of up to 50 µg /kg of PFOS for 28 days does not affect immune homeostasis in mice.


Subject(s)
Alkanesulfonic Acids , T-Lymphocytes , Humans , Mice , Animals , Cyclophosphamide/pharmacology , Alkanesulfonic Acids/pharmacology , Immunosuppressive Agents/pharmacology
5.
Chemosphere ; 307(Pt 4): 136077, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36002061

ABSTRACT

BACKGROUND: An increasing number of studies have reported that prenatal per- and polyfluoroalkyl substances (PFAS) exposure may increase childhood adiposity. However, limited data is available in China, and the overall effects of PFAS mixture remain unclear. OBJECTIVE: To examine the association of prenatal exposure to individual PFAS and their mixture with childhood adiposity at 7 years of age. METHODS: A total of 206 mother-infant pairs were recruited from the Laizhou Wan (Bay) Birth Cohort in China between 2010 and 2013. Ten PFAS were measured in maternal serum. The measurements of fat mass, body fat percentage, body mass index, waist circumference, waist-to-height ratio and overweight/obesity were used to assess adiposity in children aged 7. We fitted logistic regression, linear regression and weighted quantile sum (WQS) regression models to estimate the association of prenatal exposure to individual PFAS and their mixture with childhood adiposity. RESULTS: We found negative associations of perfluoroheptanoic acid (PFHpA) and perfluorooctane sulfonamide (PFOSA) exposure with adiposity measurements in all children. The result from the WQS model consistently revealed that the PFAS mixture was inversely related to adiposity measurements. Each quartile increase of the PFAS mixture was associated with a 1.14 kg decrease (95% CI: -2.27, -0.02) in fat mass and a 2.32% decrease (95% CI: -4.51, -0.14) in body fat. Moreover, significant sex differences were found. PFAS mixture was negatively associated with five adiposity measurements in boys, but positively associated with all adiposity measurements except body fat percentage in girls. PFOSA, PFHpA and perfluorobutanesulfonate (PFBS) with weights >0.300 were the main contributors to the overall effects observed among all children, boys and girls, respectively. CONCLUSION: This study suggests potential sex-specific associations of prenatal exposure to individual PFAS and their mixture with childhood adiposity, with the observed relationship being negative for boys but positive for girls.


Subject(s)
Alkanesulfonic Acids , Environmental Pollutants , Fluorocarbons , Prenatal Exposure Delayed Effects , Adiposity , Alkanesulfonic Acids/pharmacology , Child , Female , Fluorocarbons/pharmacology , Humans , Infant , Male , Obesity , Pregnancy , Sulfonamides/pharmacology
6.
Proc Natl Acad Sci U S A ; 118(24)2021 06 15.
Article in English | MEDLINE | ID: mdl-34099560

ABSTRACT

Allergic asthma, driven by T helper 2 cell-mediated immune responses to common environmental antigens, remains the most common respiratory disease in children. Perfluorinated chemicals (PFCs) are environmental contaminants of great concern, because of their wide application, persistence in the environment, and bioaccumulation. PFCs associate with immunological disorders including asthma and attenuate immune responses to vaccines. The influence of PFCs on the immunological response to allergens during childhood is unknown. We report here that a major PFC, perfluorooctane sulfonate (PFOS), inactivates house dust mite (HDM) to dampen 5-wk-old, early weaned mice from developing HDM-induced allergic asthma. PFOS further attenuates the asthma protective effect of the microbial product lipopolysaccharide (LPS). We demonstrate that PFOS prevents desensitization of lung epithelia by LPS, thus abolishing the latter's protective effect. A close mechanistic study reveals that PFOS specifically binds the major HDM allergen Der p1 with high affinity as well as the lipid A moiety of LPS, leading to the inactivation of both antigens. Moreover, PFOS at physiological human (nanomolar) concentrations inactivates Der p1 from HDM and LPS in vitro, although higher doses did not cause further inactivation because of possible formation of PFOS aggregates. This PFOS-induced neutralization of LPS has been further validated in primary human cell models and extended to an in vivo bacterial infection mouse model. This study demonstrates that early life exposure of mice to a PFC blunts airway antigen bioactivity to modulate pulmonary inflammatory responses, which may adversely affect early pulmonary health.


Subject(s)
Alkanesulfonic Acids/pharmacology , Antigens, Dermatophagoides/immunology , Asthma/immunology , Asthma/parasitology , Fluorocarbons/pharmacology , Hypersensitivity/immunology , Hypersensitivity/parasitology , Alkanesulfonic Acids/chemistry , Animals , Antigens, Dermatophagoides/chemistry , Asthma/complications , Asthma/genetics , Dendritic Cells/immunology , Escherichia coli , Female , Fluorocarbons/chemistry , Gene Expression Profiling , Hypersensitivity/complications , Hypersensitivity/genetics , Immunomodulation/drug effects , Immunomodulation/genetics , Lipopolysaccharides , Lung/immunology , Lung/microbiology , Lung/parasitology , Lung/pathology , Mice, Inbred BALB C , Models, Molecular , Pseudomonas aeruginosa/physiology , Pyroglyphidae/physiology
7.
Biomed Res Int ; 2021: 6625952, 2021.
Article in English | MEDLINE | ID: mdl-33880372

ABSTRACT

The liver is the primary target organ for perfluorooctane sulphonate (PFOS), a recently discovered persistent organic pollutant. However, the mechanisms mediating hepatotoxicity remain unclear. Herein, we explored the relationship between reactive oxygen species (ROS) and autophagy and apoptosis induced by PFOS in L-02 cells, which are incubated with different concentrations of PFOS (0, 50, 100, 150, 200, or 250 µmol/L) for 24 or 48 hrs at 37°C. The results indicated that PFOS exposure decreased cell activities, enhanced ROS levels in a concentration-dependent manner, decreased mitochondrial membrane potential (MMP), and induced autophagy and apoptosis. Compared with the control, 200 µmol/L PFOS increased ROS levels; enhanced the expression of Bax, cleaved-caspase-3, and LC3-II; induced autophagy; decreased MMP; and lowered Bcl-2, p62, and Bcl-2/Bax ratio. The antioxidant N-acetyl cysteine (NAC) protected MMP against PFOS-induced changes and diminished apoptosis and autophagy. Compared with 200 µmol/L PFOS treatment, NAC pretreatment reversed the increase in ROS, Bax, and cleaved-caspase-3 protein caused by PFOS, lowered the apoptosis rate increased by PFOS, and increased the levels of MMP and Bcl-2/Bax ratio decreased by PFOS. The autophagy inhibitor 3-methyladenine and chloroquine decreased apoptosis and cleaved-caspase-3 protein level and increased the Bcl-2/Bax ratio. In summary, our results suggest that ROS-triggered autophagy is involved in PFOS-induced apoptosis in L-02 cells.


Subject(s)
Alkanesulfonic Acids/pharmacology , Apoptosis , Autophagy , Embryo, Mammalian/pathology , Fluorocarbons/pharmacology , Liver/embryology , Liver/pathology , Reactive Oxygen Species/metabolism , Acetylcysteine/pharmacology , Adenine/analogs & derivatives , Adenine/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Cadaverine/pharmacology , Cell Line , Cell Survival/drug effects , Cellular Microenvironment/drug effects , Humans , Liver/drug effects , Membrane Potential, Mitochondrial/drug effects , Proteins/metabolism , Vacuoles/drug effects , Vacuoles/metabolism
8.
Immunol Lett ; 233: 31-41, 2021 05.
Article in English | MEDLINE | ID: mdl-33722553

ABSTRACT

Perfluoroctanesulfonate (PFOS) belongs to a larger family of compounds known as Per- and polyfluoroalkyl substances (PFAS). The strength of the carbon-fluorine bond makes PFOS extremely resistant to environmental degradation. Due to its persistent nature, research has been directed to elucidating possible health effects of PFOS on humans and laboratory animals. Here we have explored the effects of PFOS exposure on immune development and function in mice. We exposed adult mice to 3 and 1.5 µg/kg/day of PFOS for 2 and 4 weeks, respectively, and examined the effects of PFOS exposure on populations of T cells, B cells, and granulocytes. These doses of PFOS resulted in serum levels of approximately 100 ng/mL with no weight loss during exposure. We find that PFOS does not affect T-cell development during this time. However, while PFOS exposure reduced immune cell populations in some organs, it also led to an increase in the numbers of cells in others, suggesting possible relocalization of cells. We also examined the effect of PFOS on the response to influenza virus infection. We find that exposure to PFOS at 1.5 µg/kg/day of PFOS for 4 weeks does not affect weight loss or survival, nor is viral clearance affected. Analysis of antibody and T cell specific antiviral responses indicate that at this concentration, PFOS does not suppress the immune cell development or antigen specific immune response.


Subject(s)
Alkanesulfonic Acids/pharmacology , Cell Differentiation/drug effects , Fluorocarbons/pharmacology , Granulocytes/drug effects , Granulocytes/immunology , Immunologic Factors/pharmacology , Lymphocytes/drug effects , Lymphocytes/immunology , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , Biomarkers , Bone Marrow/drug effects , Bone Marrow/immunology , Bone Marrow/metabolism , Cell Differentiation/immunology , Female , Granulocytes/cytology , Granulocytes/metabolism , Immunophenotyping , Influenza A virus/immunology , Lymphocytes/cytology , Lymphocytes/metabolism , Male , Mice , Organ Specificity , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/virology , Spleen/cytology , Spleen/drug effects , Spleen/immunology , Spleen/metabolism , Thymus Gland/immunology , Thymus Gland/metabolism
9.
FEMS Microbiol Lett ; 367(23)2020 12 22.
Article in English | MEDLINE | ID: mdl-33242089

ABSTRACT

Anaerobic bacteria are known to produce neurotoxic methylmercury [MeHg] when elemental mercury [Hg(0)] is provided as the sole mercury source. In this study, we examined the formation of MeHg in anaerobic incubations of sediment collected from the San Jacinto River estuary (Texas, USA) amended with aqueous Hg(0) to investigate the microbial communities involved in the conversion of Hg(0) to MeHg. The results show that the addition of the methanogen inhibitor 2-bromoethanesulfonate (BES) significantly decreased MeHg production. The mercury methylation gene, hgcA, was detected in these sediments using archaeal specific primers, and 16S rRNA sequencing showed that a member of the Methanosarcinaceae family of methanogens was active. These results suggest that methanogenic archaea play an underappreciated role in the production of MeHg in estuarine sediments contaminated with Hg(0).


Subject(s)
Geologic Sediments/microbiology , Methanosarcinaceae/metabolism , Methylmercury Compounds/metabolism , Microbiota , Water Pollutants, Chemical/metabolism , Alkanesulfonic Acids/pharmacology , Anaerobiosis , Archaea/genetics , Archaea/metabolism , Estuaries , Geologic Sediments/chemistry , Mercury/metabolism , Methanosarcinaceae/genetics , Methanosarcinaceae/isolation & purification , RNA, Ribosomal, 16S/genetics
10.
Neurotoxicol Teratol ; 81: 106907, 2020.
Article in English | MEDLINE | ID: mdl-32561179

ABSTRACT

Per- and polyfluoroalkyl substances (PFAS) are present in water and >99% of human serum. They are found in brains of wildlife; however, little is known about effects on the developing brain. To determine the effects of PFAS on brain and cardiac innervation, we conducted an outdoor mesocosm experiment with Northern leopard frog larvae (Rana pipiens) exposed to control, 10 ppb perfluorooctane sulfonate (PFOS), or a PFAS mixture totaling 10 ppb that mimicked aqueous film forming foam-impacted surface water (4 ppb PFOS, 3 ppb perfluorohexane sulfonate, 1.25 ppb perfluorooctanoate, 1.25 ppb perfluorohexanoate, and 0.5 ppb perfluoro-n-pentanoate). Water was spiked with PFAS and 25 larvae (Gosner stage (GS) 25) added to each mesocosm (n = 4 mesocosms per treatment). After 30 days, we harvested eight brains per mesocosm and remaining larvae developed to GS 46 (i.e. metamorphosis) before brains and hearts were collected. Weight, length, GS, and time to metamorphosis were recorded. Brain concentrations of all five PFAS were quantified using LC/MS/MS. Dopamine and metabolites, serotonin and its metabolite, norepinephrine, γ-aminobutyric acid, and glutamate were quantified using High Performance Liquid Chromatography with electrochemical detection while acetylcholine and acetylcholinesterase activity were quantified with the Invitrogen Amplex Red Acetylcholine Assay. PFOS accumulated in the brain time- and dose-dependently. After 30 days, the mixture decreased serotonin while both PFAS treatments decreased glutamate. Interestingly, acetylcholine increased in PFAS treatments at GS 46. This research shows that developmental environmentally relevant exposure to PFAS changes neurotransmitters, especially acetylcholine.


Subject(s)
Alkanesulfonic Acids/pharmacology , Brain/drug effects , Caprylates/pharmacology , Fluorocarbons/pharmacology , Synaptic Transmission/drug effects , Animals , Environmental Exposure/analysis , Larva/metabolism , Rana pipiens/metabolism , Synaptic Transmission/physiology , Water Pollutants, Chemical/metabolism
11.
Chem Res Toxicol ; 33(6): 1478-1486, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32423201

ABSTRACT

6:2 Chlorinated polyfluorinated ether sulfonate (6:2 Cl-PFESA) possesses a similar structure to perfluorooctanesulfonate (PFOS) and is the third most important polyfluoroalkyl/perfluoroalkyl substance (PFAS) found in the general population of China. Studies have indicated that 6:2 Cl-PFESA exhibits a stronger bioaccumulative and toxicological potential than PFOS and is thus of considerable environmental concern. Here, the binding characteristics of PFOS and 6:2 Cl-PFESA to human serum albumin (HSA) were explored based on in vitro and in silico methods. In the cell uptake assays, supplementation of HSA in the culture medium hindered diffusion of PFOS and 6:2 Cl-PFESA from the medium into cells. With the addition of 0.5, 10, and 200 µM HSA in the culture medium, the PFOS concentration in cells decreased by 21.4%, 78.1%, and 92.8%, whereas the 6:2 Cl-PFESA concentration in cells decreased by 28.4%, 84.4%, and 93.9%, respectively. Although no statistically significant difference between the reduction of PFOS and 6:2 Cl-PFESA was observed with 200 µM HSA in medium, the significant decrease in cellular 6:2 Cl-PFESA than PFOS after addition of 0.5 and 10 µM HSA implied that 6:2 Cl-PFESA had a stronger binding affinity than PFOS to HSA. Ultrafiltration centrifugation also suggested that 6:2 Cl-PFESA (Kd = 16.7 µM) had a higher affinity than PFOS (Kd = 30.7 µM) to HSA, though the binding molar ratios were similar, with 1 M HSA binding to 3-4 M PFOS/6:2 Cl-PFESA. Limited proteolysis further identified the core HSA peptides that bind to PFOS (peptide II, aa 189-457) and 6:2 Cl-PFESA (peptide I, aa 39-310). Using purified core peptides, 6:2 Cl-PFESA showed a stronger binding affinity than PFOS to both peptides I and II. The binding modes indicated that the chlorine and oxygen atoms in 6:2 Cl-PFESA were likely responsible for its preferential binding to Sudlow site I than to Trp214 or Sudlow site II, with the latter being the optimal binding site for PFOS. Overall, the stronger binding affinity of 6:2 Cl-PFESA to HSA may contribute to its higher bioaccumulation potential than PFOS.


Subject(s)
Alkanesulfonic Acids/pharmacology , Ethers/pharmacology , Fluorocarbons/pharmacology , Serum Albumin, Human/metabolism , Cell Line, Tumor , Humans , Molecular Docking Simulation
12.
Anal Chem ; 92(6): 4614-4622, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32096628

ABSTRACT

Perfluoroalkyl substances (PFASs) persist and are ubiquitous in the environment. The origins of PFAS toxicity and how they specifically affect the functions of proteins remain unclear. Herein, we report that PFASs can strongly inhibit the activity of human carbonic anhydrases (hCAs), which are ubiquitous enzymes that catalyze the hydration of CO2, are abundant in the blood and organs of mammals, and involved in pH regulation, ion homeostasis, and biosynthesis. The interactions between PFASs and hCAs were investigated using stopped-flow kinetic enzyme-inhibition measurements, native mass spectrometry (MS), and ligand-docking simulations. Narrow-bore emitters in native MS with inner diameters of ∼300 nm were used to directly and simultaneously measure the dissociation constants of 11 PFASs to an enzyme, which was not possible using conventional emitters. The data from native MS and stopped-flow measurements were in excellent agreement. Of 15 PFASs investigated, eight can inhibit at least one of four hCA isozymes (I, II, IX, and XII) with submicromolar inhibition constants, including perfluorooctanoic acid, perfluorooctanesulfonamide, and perfluorooctanesulfonic acid. Some PFASs, including those with both short and long perfluoromethylene chains, can effectively inhibit at least one hCA isozyme with low nanomolar inhibition constants.


Subject(s)
Alkanesulfonic Acids/pharmacology , Caprylates/pharmacology , Carbonic Anhydrase Inhibitors/pharmacology , Carbonic Anhydrases/metabolism , Environmental Pollutants/pharmacology , Fluorocarbons/pharmacology , Alkanesulfonic Acids/analysis , Caprylates/analysis , Carbonic Anhydrase Inhibitors/analysis , Crystallography, X-Ray , Environmental Pollutants/analysis , Fluorocarbons/analysis , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Ligands , Mass Spectrometry , Models, Molecular , Particle Size , Surface Properties
13.
J Agric Food Chem ; 68(2): 633-641, 2020 Jan 15.
Article in English | MEDLINE | ID: mdl-31891488

ABSTRACT

As typical perfluorinated compounds (PFCs), perfluorooctanoic acid (PFOA) and perfluorooctane sulfonate (PFOS) have been detected in various environmental media and their toxic effects have been extensively studied. Nevertheless, it remains unclear how PFCs cause cell apoptosis in healthy hepatocytes by inducing oxidative stress at the subcellular and molecular levels. In this study, the apoptotic pathways induced by PFOA and PFOS were explored. Besides, the effects of PFCs on the structure and function of lysozyme (LYZ) were investigated. After PFOA and PFOS exposure, the cell membrane and mitochondrial membrane potential were damaged. Further, PFOA and PFOS increased intracellular Ca2+ levels to 174.41 ± 1.70 and 158.91 ± 5.94%, respectively. Ultimately, caspase-3 was activated, causing cell apoptosis. As an indirect antioxidant enzyme, the molecular structure of LYZ was destroyed after interacting with PFOA and PFOS. Both PFOA and PFOS bound to the active center of LYZ, leading to the decrease of LYZ activity to 91.26 ± 0.78 and 76.01 ± 4.86%, respectively. This study demonstrates that PFOA and PFOS inhibit LYZ function, which can reduce the body's ability to resist oxidative stress, and then lead to mitochondria-mediated apoptosis.


Subject(s)
Alkanesulfonic Acids/pharmacology , Apoptosis/drug effects , Caprylates/pharmacology , Fluorocarbons/pharmacology , Hepatocytes/drug effects , Calcium/metabolism , Caspase 3/metabolism , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Oxidative Stress/drug effects
14.
Biomed Res Int ; 2020: 8818160, 2020.
Article in English | MEDLINE | ID: mdl-33457418

ABSTRACT

Perfluorooctane sulfonate (PFOS), an environmentally persistent pollutant, has been revealed to elicit hepatic toxicity. In the current study, we investigated the protective role of grape seed proanthocyanidin extract (GSPE) against PFOS-caused steatohepatitis in mice. Animals were exposed intragastrically to PFOS (10 mg/kg/day), GSPE (150 mg/kg/day), or their combination. After 21 days of treatment, mice exposed to PFOS exhibited steatosis, oxidative stress, and inflammation in the liver. Nevertheless, simultaneous administration of GSPE resumed the declined serum hepatic enzyme activities and histological abnormalities in PFOS-exposed mice. Furthermore, GSPE supplementation reduced the contents of triglyceride (TG) and total cholesterol (TC) and expression of lipid metabolism-associated genes CD36 and fatty acid-binding protein 4 (FABP4) in the liver of mice treated with PFOS. Moreover, GSPE suppressed the generation of lipid peroxidative product malondialdehyde and restored the activity of superoxide dismutase in the liver of PFOS-exposed mice. In addition, GSPE repressed the PFOS-induced hepatic overproduction of proinflammatory cytokines interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α). Our results demonstrate that GSPE attenuates PFOS-caused steatohepatitis in mice by regulating lipid metabolism, oxidative stress, and inflammatory response.


Subject(s)
Alkanesulfonic Acids/pharmacology , Fatty Liver/drug therapy , Fluorocarbons/pharmacology , Proanthocyanidins/pharmacology , Animals , CD36 Antigens/metabolism , Cholesterol/metabolism , Fatty Acid-Binding Proteins/metabolism , Hydrogen Peroxide , Inflammation , Interleukin-6/metabolism , Lipid Metabolism/drug effects , Lipid Peroxidation , Liver/drug effects , Male , Malondialdehyde/metabolism , Mice , Oxidative Stress , Triglycerides/metabolism , Tumor Necrosis Factor-alpha/metabolism
15.
Chemosphere ; 237: 124402, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31352096

ABSTRACT

Chlorinated polyfluoroalkyl ether sulfonate (Cl-PFESA) is a novel alternative of perfluorooctane sulfonate (PFOS). While its health risks remain unknown, there is preliminary evidence of developmental toxicity. In the present study, human bone mesenchymal stem cells (hBMSCs) were used to evaluate the effects of Cl-PFESA at non-cytotoxic concentrations on molecular regulation and cellular function of stem cells compared to PFOS, perfluorohexane sulfonate (PFHxS) and perfluorooctanoic acid (PFOA). Gene profiles of hBMSCs exposed to 100 nM of Cl-PFESA and the other 3 perfluoroalkyl acids (PFAAs) correlated significantly with each other. A total of 261 genes were found to be affected by all 4 compounds. Functional annotation analysis revealed that osteoblast differentiation, ERK1/2, TGFß and calcium signalling were interfered. Moreover, DUSP mRNA and P-SMAD protein, key factors in ERK and TGFß/SMAD signaling, were decreased by Cl-PFESA. Furthermore, intracellular calcium image suggested that calcium transients were enhanced by Cl-PFESA with lower effective concentrations and more prolonged induction than PFOS and PFHxS. Immunofluorescence staining confirmed that the stemness marker CD44 was dose-dependently repressed by Cl-PFESA. In the osteogenic differentiation following exposure to 100 nM of Cl-PFESA, both mRNA and protein of RUNX2, a target of multiple osteogenic pathways, was depressed on differentiation day 7. Exposure to Cl-PFESA at human relevant concentrations during a vulnerable period before differentiation posed persistent effects on hBMSCs, with common or even stronger potency compared to PFAAs.


Subject(s)
Alkanesulfonic Acids/pharmacology , Fluorocarbons/pharmacology , Mesenchymal Stem Cells/drug effects , Alkanesulfonates/pharmacology , Caprylates/pharmacology , Cell Differentiation/drug effects , Ether , Genetic Profile , Humans , Mesenchymal Stem Cells/cytology , Osteogenesis/drug effects , Signal Transduction/drug effects
16.
Chem Res Toxicol ; 32(8): 1656-1669, 2019 08 19.
Article in English | MEDLINE | ID: mdl-31340646

ABSTRACT

Methylmercury (MeHg) and perfluorooctanesulfonate (PFOS) are major contaminants of human blood that are both common in dietary fish, thereby raising questions about their combined impact on human development. Here, pregnant Sprague-Dawley rats ingested a daily dose, from gestational day 1 through to weaning, of either 1 mg/kg bw PFOS (PFOS-only), 1 mg/kg MeHg (MeHg-only), a mixture of 0.1 mg/kg PFOS and 1 mg/kg MeHg (Low-Mix), or of 1 mg/kg of PFOS and 1 mg/kg MeHg (High-Mix). Newborns were monitored for physical milestones and reflexive developmental responses, and in juveniles the spontaneous activity, anxiety, memory, and cognition were assessed. Targeted metabolomics of 199 analytes was applied to sectioned brain regions of juvenile offspring. Newborns in the High-Mix group had decreased weight gain as well as delayed reflexes and innate behavioral responses compared to controls and individual chemical groups indicating a toxicological interaction on early development. In juveniles, cumulative mixture effects increased in a dose-dependent manner in tests of anxiety-like behavior. However, other developmental test results suggested antagonism, as PFOS-only and MeHg-only juveniles had increased hyperactivity and thigmotaxic behavior, respectively, but fewer effects in Low-Mix and High-Mix groups. Consistent with these behavioral observations, a pattern of antagonism was also observed in neurochemicals measured in rat cortex, as PFOS-only and MeHg-only juveniles had altered concentrations of metabolites (e.g., lipids, amino acids, and biogenic amines), while no changes were evident in the combined exposures. The cortical metabolites altered in PFOS-only and MeHg-only exposed groups are involved in inhibitory and excitatory neurotransmission. These proof-of-principle findings at relatively high doses indicate the potential for toxicological interaction between PFOS and MeHg, with developmental-stage specific effects. Future mixture studies at lower doses are warranted, and prospective human birth cohorts should consider possible confounding effects from PFOS and mercury exposure on neurodevelopment.


Subject(s)
Alkanesulfonic Acids/pharmacology , Behavior, Animal/drug effects , Brain/drug effects , Brain/metabolism , Fluorocarbons/pharmacology , Metabolomics , Methylmercury Compounds/pharmacology , Alkanesulfonic Acids/administration & dosage , Alkanesulfonic Acids/analysis , Animals , Brain/pathology , Dose-Response Relationship, Drug , Female , Fluorocarbons/administration & dosage , Fluorocarbons/analysis , Male , Methylmercury Compounds/administration & dosage , Methylmercury Compounds/analysis , Pregnancy , Rats , Rats, Sprague-Dawley
17.
Chem Res Toxicol ; 32(5): 935-942, 2019 05 20.
Article in English | MEDLINE | ID: mdl-31016965

ABSTRACT

Artificial environmental endocrine disrupting chemicals (EDCs) exert public health concerns. Exposure to EDCs may induce various disorders in the cardiometabolic system. However, the underlying mechanisms remain largely unknown. Over the past decade, an abundance of evidence has emerged demonstrating a close link between cardiometabolic disorders and inflammation. The aim of the present study was to evaluate the immunological effects on macrophages from six EDCs via sirtuin (SIRT) regulation using the murine macrophage RAW 264.7 cell. We studied first the effects of these EDCs, including a series of doses of benzyl butyl phthalate (BBP), bisphenol A (BPA), diethylhexyl phthalate (DEHP), mono-(2-ethylhexyl)phthalate (MEHP), perfluorooctanoate (PFOA), or perfluorooctanesulfonate (PFOS), on SIRT1-7 transcriptional level. Among these EDCs, MEHP significantly decreased all sirtuin genes' expression in a dose-dependent manner. Under MEHP treatment, SIRT activity and protein expression were significantly decreased, while the protein expression of acetylated NF-κB was significantly increased along with significant increases in IL-1ß transcription. These results indicate that MEHP may induce the inflammatory response via SIRT-mediated acetylation of NF-κB. Additionally, the enhanced IL-1ß secretion in the presence of 50 µM MEHP ( P < 0.01) also supports inflammasome activation (significant ASC and NLRP3 protein augmentation). Both events may be regulated by MEHP induced reactive oxygen species ( P < 0.01). In conclusion, our study suggests for the first time that EDCs differentially modulate sirtuins' gene expression levels in macrophages and that a specific phthalate MEHP can lead to an increased inflammatory response by impairing vital epigenetic regulators and inflammasome activation.


Subject(s)
Diethylhexyl Phthalate/analogs & derivatives , Endocrine Disruptors/pharmacology , Gene Expression Regulation/drug effects , Inflammasomes/metabolism , Inflammation/etiology , Sirtuins/metabolism , Acetylation/drug effects , Alkanesulfonic Acids/pharmacology , Animals , Benzhydryl Compounds/pharmacology , Caprylates/pharmacology , Diethylhexyl Phthalate/pharmacology , Epigenesis, Genetic/drug effects , Fluorocarbons/pharmacology , Inflammation/genetics , Interleukin-1beta/metabolism , Mice , NF-kappa B/chemistry , NF-kappa B/metabolism , Phenols/pharmacology , Phthalic Acids/pharmacology , RAW 264.7 Cells , Reactive Oxygen Species/metabolism , Sirtuins/genetics
18.
Chemosphere ; 220: 687-695, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30605811

ABSTRACT

The ubiquitous environmental obesogens tributyltin (TBT) and perfluorooctane sulfonate (PFOS) may accumulate in parent and be transferred to their offspring, resulting in trans-generational adverse effects. In this study, we investigated the combined toxic and obesogenic effects of TBT and PFOS on the early life stages of Japanese medaka (Oryzias latipes). In ovo nanoinjection was used to simulate the maternal transfer process. Doses were controlled at 0, 0.05, 0.5, and 2.5 ng/egg (TBT) and at 0, 0.05, 0.5, and 5.0 ng/egg (PFOS), with a full factorial design for mixture formulations. Relatively high doses of agents in mixtures were needed to induce significant mortality (TBT ≥ 0.5 ng/egg) or delayed hatching (PFOS = 5.0 ng/egg) of embryos. The interaction between TBT and PFOS in mixtures had significant effects on the observed hatching delay, but not on acute mortality. Compared with controls, separate exposure to TBT (or PFOS) notably elevated adipose areas at the doses of 0.05 and 0.5 ng/egg, but not at the highest doses. Combined exposure significantly promoted the fat accumulation in newly hatched larvae, even when the doses of TBT and PFOS were both at the levels that did not show obesogenic effect. The interactive effect of TBT and PFOS could aggravate the total obesogenic effect of their mixtures, indicating a synergistic interaction. These results highlight the importance of paying close attention to interaction effects when addressing the impacts of mixtures of environmental obesogens.


Subject(s)
Adipose Tissue/drug effects , Alkanesulfonic Acids/pharmacology , Fluorocarbons/pharmacology , Oryzias/metabolism , Trialkyltin Compounds/pharmacology , Adipose Tissue/growth & development , Animals , Drug Synergism , Embryo, Nonmammalian/drug effects , Female , Larva/drug effects , Maternal Exposure/adverse effects , Obesity/chemically induced , Oryzias/embryology
19.
Chemosphere ; 215: 182-188, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30317088

ABSTRACT

6:2 chlorinated polyfluorinated ether sulfonate (F-53B) has been widely applied as a mist suppressant to replace perfluorooctane sulfonate (PFOS) in the metal plating industry in China for decades. Recently, F-53B has been frequently identified in the aquatic environment and wild-caught fish. However, studies on the uptake and elimination kinetics, and the toxicological effects of F-53B were very scarce. In this study, zebrafish larvae (72 h post fertilization, hpf) were exposed to F-53B (10, 100 µg/L) for 48 h, followed by a 24 h of depuration to examine both the dynamics of accumulation and elimination of F-53B and responses of antoxidant defense system in fish. The results showed that F-53B rapidly accumulated in zebrafish larvae in a concentration and time-dependent manner with BCF values of 3612-3615, but was eliminated slowly (half-life ranged from 241.5 to 258.6 h). F-53B exposure induced oxidative stress in zebrafish larvae, as reflected by the reduction in the GSH and MDA contents, CAT, SOD, CuZn-SOD, and GSH-ST activities, and the increase in GSH-Px activity as well as CAT and SOD protein levels. However, these oxidative stress markers were restored to control levels except for a decrease in protein level of SOD after depuration. Collectively, the results of this work indicate that F-53B behaves like PFOS and is bioaccumulative and persistent in zebrafish larvae, and further induced oxidative stress responses.


Subject(s)
Alkanesulfonates/pharmacokinetics , Larva/metabolism , Oxidative Stress/drug effects , Zebrafish/metabolism , Alkanesulfonates/analysis , Alkanesulfonates/pharmacology , Alkanesulfonic Acids/pharmacology , Animals , Biomarkers/metabolism , China , Fluorocarbons/pharmacology , Larva/drug effects , Stigmasterol/analogs & derivatives , Superoxide Dismutase/metabolism , Water Pollutants, Chemical/analysis
20.
Bioresour Technol ; 274: 557-560, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30578015

ABSTRACT

Microbial electrolysis cells (MECs) for hydrogen production exhibit great advantages over many other biohydrogen production techniques in terms of versatility of substrate and hydrogen yield. However, hydrogen and acetate scavenging by methanogens puts forward a great challenge to the application of single chamber MECs when using mixed culture. In this study, we investigated the feasibility of using acetylene, a low-cost fuel and chemical building block, to selectively inhibit methanogenesis in single chamber MECs. Results demonstrate that the periodical injection of low concentration acetylene (1% and 5%) can successfully inhibit methanogenesis in MECs using both acetate and glucose as substrates. Current generation by exoelectrogens and the syntrophy between fermentative bacteria and exoelectrogens, however, were not negatively affected. Compared with the classic methanogen inhibitor, 2-Bromoethanesulfonate (BES), the low concentration acetylene demonstrates superior effectiveness in MECs. These results demonstrate the great potential of using acetylene as a cost-effective inhibitor against methanogenesis in MECs.


Subject(s)
Acetylene/metabolism , Methane/biosynthesis , Alkanesulfonic Acids/pharmacology , Bacteria/drug effects , Electrolysis/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...