Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 177
Filtrar
1.
Ecotoxicol Environ Saf ; 278: 116402, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38728940

RESUMEN

Perfluorobutanesulfonic acid (PFBS), a short-chain alternative to perfluorooctanesulfonic acid (PFOS), is widely used in various products and is increasingly present in environmental media and human bodies. Recent epidemiological findings have raised concerns about its potential adverse health effects, although the specific toxic mechanism remains unclear. This study aimed to investigate the metabolic toxicity of gestational PFBS exposure in maternal rats. Pregnant Sprague Dawley (SD) rats were randomly assigned to three groups and administered either 3% starch gel (control), 5, or 50 mg/kg bw·d PFBS. Oral glucose tolerance tests (OGTT) and lipid profiles were measured, and integrated omics analysis (transcriptomics and non-targeted metabolomics) was employed to identify changes in genes and metabolites and their relationships with metabolic phenotypes. The results revealed that rats exposed to 50 mg/kg bw·d PFBS exhibited a significant decrease in 1-h glucose levels and the area under the curve (AUC) of OGTT compared with the starch group. Transcriptomics analysis indicated significant alterations in gene expression related to cytochrome P450 exogenous metabolism, glutathione metabolism, bile acid secretion, tumor pathways, and retinol metabolism. Differentially expressed metabolites (DEMs) were enriched in pathways such as pyruvate metabolism, the glucagon signaling pathway, central carbon metabolism in cancer, and the citric acid cycle. Co-enrichment analysis and pairwise correlation analysis among genes, metabolites, and outcomes identified several differentially expressed genes (DEGs), including Gstm1, Kit, Adcy1, Gck, Ppp1r3c, Ppp1r3d, and DEMs such as fumaric acid, L-lactic acid, 4-hydroxynonenal, and acetylvalerenolic acid. These DEGs and DEMs may play a role in the modulation of glucolipid metabolic pathways. In conclusion, our results suggest that gestational exposure to PFBS may induce molecular perturbations in glucose homeostasis. These findings provide insights into the potential mechanisms contributing to the heightened risk of abnormal glucose tolerance associated with PFBS exposure.


Asunto(s)
Fluorocarburos , Homeostasis , Ratas Sprague-Dawley , Animales , Femenino , Embarazo , Fluorocarburos/toxicidad , Ratas , Homeostasis/efectos de los fármacos , Glucosa/metabolismo , Ácidos Sulfónicos/toxicidad , Prueba de Tolerancia a la Glucosa , Metabolómica , Contaminantes Ambientales/toxicidad , Glucemia , Exposición Materna/efectos adversos , Multiómica
2.
Ecotoxicol Environ Saf ; 279: 116501, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38805831

RESUMEN

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


Asunto(s)
Desarrollo Óseo , Lactancia , Exposición Materna , Ratones Endogámicos C57BL , Efectos Tardíos de la Exposición Prenatal , Animales , Femenino , Masculino , Embarazo , Ratones , Exposición Materna/efectos adversos , Desarrollo Óseo/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Fluorocarburos/toxicidad , Osteoprotegerina/metabolismo , Osteoclastos/efectos de los fármacos , Huesos/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Ácidos Sulfónicos/toxicidad
3.
Toxicol Sci ; 200(1): 57-69, 2024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-38603627

RESUMEN

Per- and polyfluoroalkyl substances (PFAS) are a group of synthetic chemicals that are resistant to biodegradation and are environmentally persistent. PFAS are found in many consumer products and are a major source of water and soil contamination. This study investigated the effects of an environmentally relevant PFAS mixture (perfluorooctanoic acid [PFOA], perfluorooctanesulfonic acid [PFOS], perfluorohexanesulfonic acid [PFHxS]) on the transcriptome and function of human granulosa cells (hGCs). Primary hGCs were harvested from follicular aspirates of healthy, reproductive-age women who were undergoing oocyte retrieval for in vitro fertilization. Liquid Chromatography with tandem mass spectrometry (LC/MS-MS) was performed to identify PFAS compounds in pure follicular fluid. Cells were cultured with vehicle control or a PFAS mixture (2 nM PFHxS, 7 nM PFOA, 10 nM PFOS) for 96 h. Analyses of cell proliferation/apoptosis, steroidogenesis, and gene expression were measured via 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assays/immunofluorescence, ELISA/western blotting, and RNA sequencing/bioinformatics, respectively. PFOA, PFOS, and PFHxS were detected in 100% of follicle fluid samples. Increased cell proliferation was observed in hGCs treated with the PFAS mixture with no impacts on cellular apoptosis. The PFAS mixture also altered steroid hormone synthesis, increasing both follicle-stimulating hormone-stimulated and basal progesterone secretion and concomitant upregulation of STAR protein. RNA sequencing revealed inherent differences in transcriptomic profiles in hGCs after PFAS exposure. This study demonstrates functional and transcriptomic changes in hGCs after exposure to a PFAS mixture, improving our knowledge about the impacts of PFAS exposures and female reproductive health. These findings suggest that PFAS compounds can disrupt normal granulosa cell function with possible long-term consequences on overall reproductive health.


Asunto(s)
Ácidos Alcanesulfónicos , Caprilatos , Proliferación Celular , Fluorocarburos , Células de la Granulosa , Humanos , Femenino , Fluorocarburos/toxicidad , Células de la Granulosa/efectos de los fármacos , Células de la Granulosa/metabolismo , Proliferación Celular/efectos de los fármacos , Ácidos Alcanesulfónicos/toxicidad , Caprilatos/toxicidad , Células Cultivadas , Contaminantes Ambientales/toxicidad , Transcripción Genética/efectos de los fármacos , Transcriptoma/efectos de los fármacos , Ácidos Sulfónicos/toxicidad , Líquido Folicular/metabolismo , Adulto , Hormonas Esteroides Gonadales/biosíntesis , Hormonas Esteroides Gonadales/metabolismo
4.
Food Chem Toxicol ; 186: 114578, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38458531

RESUMEN

Per- and polyfluoroalkyl substances (PFAS) are a large group of stable synthetic surfactants that are incorporated into numerous products for their water and oil resistance and have been associated with adverse health effects. The present study evaluated the systemic and immunotoxicity of sub-chronic 28- or 10-day dermal exposure of PFHxS (0.625-5% or 15.63-125 mg/kg/dose) in a murine model. Elevated levels of PFHxS were detected in the serum and urine, suggesting that absorption is occurring through the dermal route. Liver weight (% body) significantly increased and spleen weight (% body) significantly decreased with PFHxS exposure, which was supported by histopathological changes. Additionally, PFHxS significantly reduced the humoral immune response and altered immune subsets in the spleen, suggesting immunosuppression. Gene expression changes were observed in the liver, skin, and spleen with genes involved in fatty acid metabolism, necrosis, and inflammation. Immune-cell phenotyping identified significant decreases in B-cells, NK cells, and CD11b+ monocyte/macrophages in the spleen along with increases in CD4+ and CD8+ T-cells, NK cells, and neutrophils in the skin. These findings support dermal PFHxS-induced liver damage and immune suppression. Overall, data support PFHxS absorption through the skin and demonstrate immunotoxicity via this exposure route, suggesting the need for further examination.


Asunto(s)
Ácidos Alcanesulfónicos , Contaminantes Ambientales , Fluorocarburos , Ratones , Animales , Modelos Animales de Enfermedad , Linfocitos T CD8-positivos , Ácidos Sulfónicos/toxicidad , Fluorocarburos/análisis
5.
Environ Sci Technol ; 57(10): 4199-4207, 2023 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-36854060

RESUMEN

Perfluoroethylcyclohexanesulfonate (PFECHS) is an emerging perfluoroalkyl substance (PFAS) that has been considered a potential replacement for perfluorooctanesulfonic acid (PFOS). However, there is little information characterizing the toxic potency of PFECHS to zebrafish embryos and its potential for effects in aquatic environments. This study assessed toxic potency of PFECHS in vivo during both acute (96-hour postfertilization) and chronic (21-day posthatch) exposures and tested concentrations of PFECHS from 500 ng/L to 2 mg/L. PFECHS was less likely to cause mortalities than PFOS for both the acute and chronic experiments based on previously published values for PFOS exposure, but exposure resulted in a similar incidence of deformities. Exposure to PFECHS also resulted in significantly increased abundance of transcripts of peroxisome proliferator activated receptor alpha (pparα), cytochrome p450 1a1 (cyp1a1), and apolipoprotein IV (apoaIV) at concentrations nearing those of environmental relevance. Overall, these results provide further insight into the safety of an emerging PFAS alternative in the aquatic environment and raise awareness that previously considered "safer" alternatives may show similar effects as legacy PFASs.


Asunto(s)
Ácidos Alcanesulfónicos , Fluorocarburos , Contaminantes Químicos del Agua , Animales , Pez Cebra , Ácidos Sulfónicos/toxicidad , Ácidos Alcanesulfónicos/toxicidad , Fluorocarburos/toxicidad , Fluorocarburos/análisis , Contaminantes Químicos del Agua/toxicidad , Contaminantes Químicos del Agua/análisis
6.
Arch Toxicol ; 96(11): 3127-3139, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35976416

RESUMEN

This commentary proposes an approach to risk assessment of mixtures of per- and polyfluorinated alkyl substances (PFAS) as EFSA was tasked to derive a tolerable intake for a group of 27 PFAS. The 27 PFAS to be considered contain different functional groups and have widely variable physicochemical (PC) properties and toxicokinetics and thus should not treated as one group based on regulatory guidance for risk assessment of mixtures. The proposed approach to grouping is to split the 27 PFAS into two groups, perfluoroalkyl carboxylates and perfluoroalkyl sulfonates, and apply a relative potency factor approach (as proposed by RIVM) to obtain two separate group TDIs based on liver toxicity in rodents since liver toxicity is a sensitive response of rodents to PFAS. Short chain PFAS and other PFAS structures should not be included in the groups due to their low potency and rapid elimination. This approach is in better agreement with scientific and regulatory guidance for mixture risk assessment.


Asunto(s)
Ácidos Alcanesulfónicos , Fluorocarburos , Ácidos Alcanesulfónicos/toxicidad , Ácidos Carboxílicos/toxicidad , Fluorocarburos/química , Fluorocarburos/toxicidad , Medición de Riesgo , Ácidos Sulfónicos/toxicidad
7.
Chemosphere ; 287(Pt 2): 132121, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34509758

RESUMEN

Since the voluntary phaseout of perfluorooctanesulfonic acid (PFOS), smaller congeners, such as perfluorobutanesulfonic acid (PFBS) have served as industrial replacements and been detected in contaminated aquifers. This study sought to examine the effects of a maternal preconception PFBS exposure on the development of eggs and healthy offspring. Adult female zebrafish received a one-week waterborne exposure of 0.08, 0.14, and 0.25 mg/L PFBS. After which, females were bred with non-exposed males and embryos collected over 5 successful breeding events. PFBS concentrations were detected in levels ranging from 99 to 253 pg/embryo in the first collection but were below the limit of quantitation by fourth and fifth clutches. Therefore, data were subsequently binned into early collection embryos in which PFBS was detected and late collections, in which PFBS was below quantitation. In the early collection, embryo 24 h survival was significantly reduced. In the late collection, embryo development was impacted with unique patterns emerging between Nrf2a wildtype and mutant larvae. Additionally, the impact of nutrient loading into the embryos was assessed through measurement of fatty acid profiles, total cholesterol, and triglyceride content. There were no clear dose-dependent effects, but again unique patterns were observed between the genotypes. Preconception PFBS exposures were found to alter egg and embryo development, which is mediated by direct toxicant loading in the eggs, nutrient loading into eggs, and the function of Nrf2a. These findings provide insight into the reproductive and developmental effects of PFBS and identify maternal preconception as a novel critical window of exposure.


Asunto(s)
Fluorocarburos , Pez Cebra , Animales , Desarrollo Embrionario , Femenino , Fluorocarburos/toxicidad , Humanos , Masculino , Exposición Materna , Ácidos Sulfónicos/toxicidad , Pez Cebra/genética
8.
Toxicology ; 465: 153024, 2022 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-34743024

RESUMEN

Human exposure to per- and polyfluoroalkyl substances (PFAS) is ubiquitous, with mixtures of PFAS detected in drinking water, food, household dust, and other exposure sources. Animal toxicity studies and human epidemiology indicate that PFAS may act through shared mechanisms including activation of peroxisome proliferator activated receptor α (PPARα). However, the effect of PFAS mixtures on human relevant molecular initiating events remains an important data gap in the PFAS literature. Here, we tested the ability of modeling approaches to predict the effect of diverse PPARα ligands on receptor activity using Cos7 cells transiently transfected with a full length human PPARα (hPPARα) expression construct and a peroxisome proliferator response element-driven luciferase reporter. Cells were treated for 24 h with two full hPPARα agonists (pemafibrate and GW7647), a full and a partial hPPARα agonist (pemafibrate and mono(2-ethylhexyl) phthalate), or a full hPPARα agonist and a competitive antagonist (pemafibrate and GW6471). Receptor activity was modeled with three additive approaches: effect summation, relative potency factors (RPF), and generalized concentration addition (GCA). While RPF and GCA accurately predicted activity for mixtures of full hPPARα agonists, only GCA predicted activity for full and partial hPPARα agonists and a full agonist and antagonist. We then generated concentration response curves for seven PFAS, which were well-fit with three-parameter Hill functions. The four perfluorinated carboxylic acids (PFCA) tended to act as full hPPARα agonists while the three perfluorinated sulfonic acids (PFSA) tended to act as partial agonists that varied in efficacy between 28-67 % of the full agonist, positive control level. GCA and RPF performed equally well at predicting the effects of mixtures with three PFCAs, but only GCA predicted experimental activity with mixtures of PFSAs and a mixture of PFCAs and PFSAs at ratios found in the general population. We conclude that of the three approaches, GCA most accurately models the effect of PFAS mixtures on hPPARα activity in vitro. Understanding the differences in efficacy with which PFAS activate hPPARα is essential for accurately predicting the effects of PFAS mixtures. As PFAS can activate multiple nuclear receptors, future analyses should examine mixtures effects in intact cells where multiple molecular initiating events contribute to proximate effects and functional changes.


Asunto(s)
Ácidos Carboxílicos/toxicidad , Hidrocarburos Fluorados/toxicidad , Modelos Moleculares , PPAR alfa/agonistas , PPAR alfa/antagonistas & inhibidores , Ácidos Sulfónicos/toxicidad , Animales , Células COS , Chlorocebus aethiops , Relación Dosis-Respuesta a Droga , Agonismo Parcial de Drogas , Estructura Molecular , PPAR alfa/genética , PPAR alfa/metabolismo , Transducción de Señal , Relación Estructura-Actividad
9.
Nutrients ; 13(11)2021 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-34836157

RESUMEN

Per- and polyfluoroalkyl substances (PFAS) are synthetic chemicals utilized in various industrial settings and include products such as flame retardants, artificial film-forming foams, cosmetics, and non-stick cookware, among others. Epidemiological studies suggest a link between increased blood PFAS levels and prostate cancer incidence, but the mechanism through which PFAS impact cancer development is unclear. To investigate the link between PFAS and prostate cancer, we evaluated the impact of metabolic alterations resulting from a high-fat diet combined with PFAS exposure on prostate tumor progression. We evaluated in vivo prostate cancer xenograft models exposed to perfluorooctane sulfonate (PFOS), a type of PFAS compound, and different diets to study the effects of PFAS on prostate cancer progression and metabolic activity. Metabolomics and transcriptomics were used to understand the metabolic landscape shifts upon PFAS exposure. We evaluated metabolic changes in benign or tumor cells that lead to epigenomic reprogramming and altered signaling, which ultimately increase tumorigenic risk and tumor aggressiveness. Our studies are the first in the field to provide new and clinically relevant insights regarding novel metabolic and epigenetic states as well as to support the future development of effective preventative and therapeutic strategies for PFAS-induced prostate cancers. Our findings enhance understanding of how PFAS synergize with high-fat diets to contribute to prostate cancer development and establish an important basis to mitigate PFAS exposure.


Asunto(s)
Ácidos Alcanesulfónicos/toxicidad , Dieta Alta en Grasa , Fluorocarburos/toxicidad , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Ácidos Sulfónicos/toxicidad , Acetilación , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Progresión de la Enfermedad , Xenoinjertos , Histonas/metabolismo , Humanos , Masculino , Ratones , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Transducción de Señal/efectos de los fármacos
10.
J Med Chem ; 64(16): 12322-12358, 2021 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-34378914

RESUMEN

Rhodesain is a major cysteine protease of Trypanosoma brucei rhodesiense, a pathogen causing Human African Trypanosomiasis, and a validated drug target. Recently, we reported the development of α-halovinylsulfones as a new class of covalent reversible cysteine protease inhibitors. Here, α-fluorovinylsulfones/-sulfonates were optimized for rhodesain based on molecular modeling approaches. 2d, the most potent and selective inhibitor in the series, shows a single-digit nanomolar affinity and high selectivity toward mammalian cathepsins B and L. Enzymatic dilution assays and MS experiments indicate that 2d is a slow-tight binder (Ki = 3 nM). Furthermore, the nonfluorinated 2d-(H) shows favorable metabolism and biodistribution by accumulation in mice brain tissue after intraperitoneal and oral administration. The highest antitrypanosomal activity was observed for inhibitors with an N-terminal 2,3-dihydrobenzo[b][1,4]dioxine group and a 4-Me-Phe residue in P2 (2e/4e) with nanomolar EC50 values (0.14/0.80 µM). The different mechanisms of reversible and irreversible inhibitors were explained using QM/MM calculations and MD simulations.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Inhibidores de Cisteína Proteinasa/farmacología , Sulfonas/farmacología , Ácidos Sulfónicos/farmacología , Tripanocidas/farmacología , Compuestos de Vinilo/farmacología , Animales , Cisteína Endopeptidasas/química , Inhibidores de Cisteína Proteinasa/síntesis química , Inhibidores de Cisteína Proteinasa/metabolismo , Inhibidores de Cisteína Proteinasa/toxicidad , Pruebas de Enzimas , Femenino , Células HeLa , Humanos , Cinética , Masculino , Ratones , Simulación del Acoplamiento Molecular , Estructura Molecular , Pruebas de Sensibilidad Parasitaria , Unión Proteica , Relación Estructura-Actividad , Sulfonas/síntesis química , Sulfonas/metabolismo , Sulfonas/toxicidad , Ácidos Sulfónicos/síntesis química , Ácidos Sulfónicos/metabolismo , Ácidos Sulfónicos/toxicidad , Tripanocidas/síntesis química , Tripanocidas/metabolismo , Tripanocidas/toxicidad , Trypanosoma brucei brucei/efectos de los fármacos , Compuestos de Vinilo/síntesis química , Compuestos de Vinilo/metabolismo , Compuestos de Vinilo/toxicidad
11.
Toxicology ; 462: 152921, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34464680

RESUMEN

Per- and polyfluoroalkyl substances (PFAS) are a family of chemicals that are ubiquitous in the environment. Some of these chemicals, such as perfluorooctanesulfonic acid (PFOS), perfluorohexanesulfonate (PFHxS) and perfluorooctanoic acid (PFOA), are found in human sera and have been shown to cause liver steatosis and reduce postnatal survival and growth in rodents. The purpose of this work is to evaluate the impact of diet and PFAS exposure to mouse dam (mus musculus) on the risk to pup liver and metabolism endpoints later in life, as well as evaluate PFAS partitioning to pups. Timed-pregnant dams were fed a standard chow diet or 60 % kcal high fat diet (HFD). Dams were administered either vehicle, 1 mg/kg PFOA, 1 mg/kg PFOS, 1 mg/kg PFHxS, or a PFAS mixture (1 mg/kg of each PFOA, PFOS, and PFHxS) daily via oral gavage from gestation day 1 until postnatal day (PND) 20. At PND 21, livers of dams and 2 pups of each sex were evaluated for lipid changes while remaining pups were weaned to the same diet as the dam for an additional 10 weeks. Dam and pup serum at PND 21 and PND 90 were also evaluated for PFAS concentration, alanine aminotransferase (ALT), leptin and adiponectin, and glycosylated hemoglobin A1c. Perinatal exposure to a HFD, as expected, increased pup body weight, maternal liver weight, pup liver triglycerides, pup serum ALT, and pup serum leptin. PFOA and the PFAS mixture increased liver weights, and. treatment with all three compounds increased liver triglycerides. The maternal HFD increased dam and pup serum PFAS levels, however, was protective against PFOA-induced increase in serum ALT and observed increases in liver triglycerides. The PFAS mixture had very distinct effects when compared to single compound treatment, suggesting some cumulative effects, particularly when evaluating PFAS transfer from dam to pup. This data highlights the importance of diet and mixtures when evaluating liver effect of PFAS and PFAS partitioning.


Asunto(s)
Ácidos Alcanesulfónicos/toxicidad , Caprilatos/toxicidad , Dieta Alta en Grasa/efectos adversos , Fluorocarburos/toxicidad , Ácidos Sulfónicos/toxicidad , Animales , Contaminantes Ambientales/toxicidad , Femenino , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Exposición Materna/efectos adversos , Ratones , Embarazo , Efectos Tardíos de la Exposición Prenatal/fisiopatología
12.
Environ Health ; 20(1): 76, 2021 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-34193151

RESUMEN

BACKGROUND: Per- and polyfluoroalkyl substances (PFAS) and polybrominated diphenyl ethers (PBDEs) are endocrine disrupting chemicals with widespread exposures across the U.S. given their abundance in consumer products. PFAS and PBDEs are associated with reproductive toxicity and adverse health outcomes, including certain cancers. PFAS and PBDEs may affect health through alternations in telomere length. In this study, we examined joint associations between prenatal exposure to PFAS, PBDEs, and maternal and newborn telomere length using mixture analyses, to characterize effects of cumulative environmental chemical exposures. METHODS: Study participants were enrolled in the Chemicals in Our Bodies (CIOB) study, a demographically diverse cohort of pregnant people and children in San Francisco, CA. Seven PFAS (ng/mL) and four PBDEs (ng/g lipid) were measured in second trimester maternal serum samples. Telomere length (T/S ratio) was measured in delivery cord blood of 292 newborns and 110 second trimester maternal whole blood samples. Quantile g-computation was used to assess the joint associations between groups of PFAS and PBDEs and newborn and maternal telomere length. Groups considered were: (1) all PFAS and PBDEs combined, (2) PFAS, and (3) PBDEs. Maternal and newborn telomere length were modeled as separate outcomes. RESULTS: T/S ratios in newborn cord and maternal whole blood were moderately correlated (Spearman ρ = 0.31). In mixtures analyses, a simultaneous one quartile increase in all PFAS and PBDEs was associated with a small increase in newborn (mean change per quartile increase = 0.03, 95% confidence interval [CI] = -0.03, 0.08) and maternal telomere length (mean change per quartile increase = 0.03 (95% CI = -0.03, 0.09). When restricted to maternal-fetal paired samples (N = 76), increasing all PFAS and PBDEs combined was associated with a strong, positive increase in newborn telomere length (mean change per quartile increase = 0.16, 95% CI = 0.03, 0.28). These associations were primarily driven by PFAS (mean change per quartile increase = 0.11 [95% CI = 0.01, 0.22]). No associations were observed with maternal telomere length among paired samples. CONCLUSIONS: Our findings suggest that PFAS and PBDEs may be positively associated with newborn telomere length.


Asunto(s)
Contaminantes Ambientales/toxicidad , Retardadores de Llama/toxicidad , Fluorocarburos/toxicidad , Éteres Difenilos Halogenados/toxicidad , Efectos Tardíos de la Exposición Prenatal , Telómero/efectos de los fármacos , Adulto , Monitoreo Biológico , Contaminantes Ambientales/análisis , Ácidos Grasos/análisis , Ácidos Grasos/toxicidad , Femenino , Retardadores de Llama/análisis , Fluorocarburos/análisis , Éteres Difenilos Halogenados/análisis , Humanos , Recién Nacido , Masculino , Exposición Materna , Intercambio Materno-Fetal , Embarazo , Ácidos Sulfónicos/análisis , Ácidos Sulfónicos/toxicidad
13.
J Hazard Mater ; 419: 126361, 2021 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-34157464

RESUMEN

Per and poly-fluoroalkyl substances (PFAS) have been recognized as contaminants of emerging concerns by the United States Environmental Protection Agency (US EPA) due to their environmental impact. Several advisory guidelines were proposed worldwide aimed at limiting their occurrences in the aquatic environments, especially for perfluorooctane sulfonic acid (PFOS) and perfluorooctanoic acid (PFOA). This review paper aims to provide a holistic review in the emerging area of PFAS research by summarizing the spatiotemporal variations in PFAS concentrations in surface water systems globally, highlighting the possible trends of occurrences of PFAS, and presenting potential human health impacts as a result of PFAS exposure through surface water matrices. From the data analysis in this study, occurrences of PFOA and PFOS in many surface water matrices were observed to be several folds higher than the US EPA health advisory level of 70 ng/L for lifetime exposure from drinking water. Direct discharge and atmospheric deposition were identified as primary sources of PFAS in surface water and cryosphere, respectively. While global efforts focused on limiting usages of long-chain PFAS such as PFOS and PFOA, the practices of using short-chain PFAS such as perfluorobutanoic acid (PFBA) and perfluorobutane sulfonic acid (PFBS) and PFAS alternatives increased substantially. These compounds are also potentially associated with adverse impacts on human health, animals and biota.


Asunto(s)
Ácidos Alcanesulfónicos , Agua Potable , Fluorocarburos , Contaminantes Químicos del Agua , Ácidos Alcanesulfónicos/toxicidad , Animales , Agua Potable/análisis , Fluorocarburos/análisis , Fluorocarburos/toxicidad , Humanos , Ácidos Sulfónicos/toxicidad , Estados Unidos , United States Environmental Protection Agency , Contaminantes Químicos del Agua/análisis , Contaminantes Químicos del Agua/toxicidad
14.
Kaohsiung J Med Sci ; 37(9): 812-818, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34042266

RESUMEN

Ulcerative colitis (UC) is a chronic disease characterized by mucosal and submucosal inflammation, which has a low cure rate and is prone to relapse, due to the immune imbalance of the body. Inhibition of inflammation-related pathways can delay the progression of UC. Toll-like receptor 4 (TLR4) pathway is considered to be one of the important signaling pathways involved in colon inflammation. Eriodictyol (EDT) is a natural flavonoid widely distributed in foodborne plants. EDT plays an important role in the regulation of inflammation and related signaling pathways. However, whether EDT plays a role in UC remains unknown. Herein, we established a TNBS induced animal model of enteritis in Wistar rats. Our data confirmed the establishment of TNBS induced animal model of enteritis and the administration Eriodictyol in Wistar rats. EDT treatment alleviated TNBS-induced intestinal tissue injury in rats. We further found that EDT reduced MPO expression and regulated the cytokine parameters in TNBS-induced intestinal tissues of rats. The levels of TNF-α, IL-1ß, IL-6, IL-10, IL-2, and IL-12 were also affected by the treatment of EDT. EDT also affected SOD, CAT, GSH-Px, and MDA level in rats with colitis. Moreover, EDT regulated TNBS-induced TLR4/NF-κB pathway activation, therefore inhibiting the progression of UC. Our results suggest that EDT could be a potential therapeutic agent for UC.


Asunto(s)
Colitis Ulcerosa/prevención & control , Flavanonas/farmacología , FN-kappa B/metabolismo , Transducción de Señal/efectos de los fármacos , Ácidos Sulfónicos/toxicidad , Receptor Toll-Like 4/metabolismo , Animales , Colitis Ulcerosa/inducido químicamente , Ratas , Ratas Wistar
15.
J Clin Endocrinol Metab ; 106(8): 2495-2504, 2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-33890111

RESUMEN

OBJECTIVE: To investigate the prospective associations of life-course perfluoroalkyl substances (PFASs) exposure with glucose homeostasis at adulthood. METHODS: We calculated insulin sensitivity and beta-cell function indices based on 2-h oral glucose tolerance tests at age 28 in 699 Faroese born in 1986-1987. Five major PFASs were measured in cord whole blood and in serum from ages 7, 14, 22, and 28 years. We evaluated the associations with glucose homeostasis measures by PFAS exposures at different ages using multiple informant models fitting generalized estimating equations and by life-course PFAS exposures using structural equation models. RESULTS: Associations were stronger for perfluorooctane sulfonate (PFOS) and suggested decreased insulin sensitivity and increased beta-cell function-for example, ß (95% CI) for log-insulinogenic index per PFOS doubling = 0.12 (0.02, 0.22) for prenatal exposures, 0.04 (-0.10, 0.19) at age 7, 0.07 (-0.07, 0.21) at age 14, 0.05 (-0.04, 0.15) at age 22, and 0.04 (-0.03, 0.11) at age 28. Associations were consistent across ages (P for age interaction > 0.10 for all PFASs) and sex (P for sex interaction > 0.10 for all PFASs, except perfluorodecanoic acid). The overall life-course PFOS exposure was also associated with altered glucose homeostasis (P = 0.04). Associations for other life-course PFAS exposures were nonsignificant. CONCLUSIONS: Life-course PFAS exposure is associated with decreased insulin sensitivity and increased pancreatic beta-cell function in young adults.


Asunto(s)
Glucemia/metabolismo , Exposición a Riesgos Ambientales , Contaminantes Ambientales/toxicidad , Resistencia a la Insulina/fisiología , Células Secretoras de Insulina/efectos de los fármacos , Adolescente , Adulto , Ácidos Alcanesulfónicos/sangre , Ácidos Alcanesulfónicos/toxicidad , Caprilatos/sangre , Caprilatos/toxicidad , Niño , Ácidos Decanoicos/sangre , Ácidos Decanoicos/toxicidad , Contaminantes Ambientales/sangre , Ácidos Grasos , Femenino , Fluorocarburos/sangre , Fluorocarburos/toxicidad , Prueba de Tolerancia a la Glucosa , Humanos , Células Secretoras de Insulina/metabolismo , Masculino , Ácidos Sulfónicos/sangre , Ácidos Sulfónicos/toxicidad , Adulto Joven
16.
Ecotoxicol Environ Saf ; 214: 112107, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33667734

RESUMEN

Perfluorobutanesulfonate (PFBS) pollutant and probiotic bacteria can interact to affect the reproductive outcomes of zebrafish. However, it is still unexplored how the growth and health of offspring are modulated by the combination of PFBS and probiotic. In the present study, adult zebrafish were exposed to 0 and 10 µg/L PFBS for 40 days, with or without dietary supplementation of probiotic Lactobacillus rhamnosus. After parental exposure, the development, growth and viability of offspring larvae were examined, with the integration of molecular clues across proteome fingerprint, growth hormone/insulin-like growth factor (GH/IGF) axis, calcium homeostasis, hypothalamic-pituitary-adrenal (HPA) axis and nutrient metabolism. Parental probiotic supplementation significantly increased the body weight and body length of offspring larvae. Despite the spiking of PFBS, larvae from the combined exposure group still had longer body length. RNA processing and ribosomal assembly pathways may underlie the enhancement of offspring growth by probiotic bacteria. However, the presence of PFBS remarkably increased the concentrations of cortisol hormone in offspring larvae as means to cope with the xenobiotic stress, which required more energy production. As evidenced by the proteomic analysis, the addition of probiotic bacteria likely alleviated the energy metabolism disorders of PFBS, thus allocating more energy for the larval offspring growth from the combined group. It was noteworthy that multiple molecular disturbances caused by PFBS were antagonized by probiotic additive. Overall, the present study elucidated the intergenerational interaction between PFBS and probiotic on offspring growth and health after parental exposure.


Asunto(s)
Fluorocarburos/toxicidad , Lacticaseibacillus rhamnosus , Larva/efectos de los fármacos , Probióticos/farmacología , Ácidos Sulfónicos/toxicidad , Contaminantes Químicos del Agua/toxicidad , Pez Cebra/crecimiento & desarrollo , Animales , Metabolismo Energético/efectos de los fármacos , Femenino , Sistema Hipotálamo-Hipofisario , Larva/crecimiento & desarrollo , Masculino , Proteómica
17.
Toxicol Lett ; 339: 78-87, 2021 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-33387635

RESUMEN

Obesity is a complex disease with many causes, including a possible role for environmental chemicals. Perfluorohexane sulfonate (PFHxS) is one of many per- and polyfluoroalkyl substances (PFASs) frequently detected in humans and it is suspected to be an obesogenic compound. We examined the potential long-term effects of PFHxS on metabolic parameters in rats after developmental exposure to 0.05, 5 or 25 mg/kg bw/day, with or without co-exposure to a background mixture of twelve endocrine disrupting chemicals (EDmix). Both male and female offspring showed signs of lower birth weight following intrauterine exposure. Female offspring exposed to both PFHxS and EDmix had increased body weight in adulthood. The retroperitoneal fat pad was larger in the PFHxS-exposed female offspring when compared to those exposed to EDmix alone. An attempt to detect putative molecular markers in the fat tissue by performing whole transcriptome profiling revealed no significant changes between groups and there were no significant effects on plasma leptin levels in exposed females. Our results show that early life exposure to endocrine disrupting chemicals can influence body weight later in life, but the effect is not necessarily reflected in changed gene expression in the fat tissue.


Asunto(s)
Tejido Adiposo/efectos de los fármacos , Disruptores Endocrinos/metabolismo , Disruptores Endocrinos/toxicidad , Obesidad/inducido químicamente , Ácidos Sulfónicos/metabolismo , Ácidos Sulfónicos/toxicidad , Aumento de Peso/efectos de los fármacos , Adipocitos/metabolismo , Adulto , Animales , Exposición a Riesgos Ambientales/efectos adversos , Femenino , Fluorocarburos , Humanos , Masculino , Embarazo , Efectos Tardíos de la Exposición Prenatal , Ratas
18.
Toxicol In Vitro ; 70: 105011, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33038467

RESUMEN

Perfluorohexane sulfonate (PFHxS) is one of the most abundant perfluorinated compounds in the environment. Exposure to this compound has been correlated to a decrease in human fertility, although the molecular and cellular mechanisms underlying this correlation have not been described. The adverse reproductive effects of PFHxS could be based on alterations in oocyte maturation, the process rendering oocytes competent for fertilization. The aim of this study was to evaluate the effect of PFHxS on porcine oocyte viability and maturation in vitro, as well as on gap-junctional intercellular communication (GJIC) in cumulus-oocyte complexes (COCs), oocyte mitochondrial membrane potential (mΔΨ) and DNA damage in cumulus cells, as possible mechanisms of action. PFHxS caused cytotoxicity (medium lethal concentration, LC50 = 329.1 µM) and inhibition of oocyte maturation (medium inhibitory concentration, MIC50 = 91.68 µM). GJIC was not affected in exposed COCs. However, the mitochondrial membrane potential was significantly decreased in PFHxS-exposed oocytes at the germinal vesicle breakdown (GVBD) stage. In addition, exposure to PFHxS induced DNA damage in cumulus cells. Thus, inhibition of oocyte maturation by PFHxS could be attributed to a decreased oocyte mΔΨ at the GVBD and to DNA damage of the cumulus cells that support the oocyte.


Asunto(s)
Células del Cúmulo/efectos de los fármacos , Ácidos Sulfónicos/toxicidad , Animales , Comunicación Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Células del Cúmulo/fisiología , Daño del ADN , Femenino , Fluorocarburos , Uniones Comunicantes/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Oocitos/efectos de los fármacos , Oocitos/fisiología , Porcinos
19.
J Toxicol Environ Health A ; 84(3): 125-136, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33143551

RESUMEN

Perfluoroalkyl acids (PFAAs) are persistent environmental contaminants that are associated with various adverse health outcomes. Perfluorooctanoic acid (PFOA) is one of the most prominently detected PFAAs in the environment, which is now replaced with shorter chain carbon compounds including perfluorohexanoic acid (PFHxA) and perfluorobutyric acid (PFBA). The aim of this study was to compare the toxicity of four PFAAs as a function of chain length and head group (carboxylate versus sulfonate) with in vitro and in vivo zebrafish assessments, which were subsequently compared to other cell and aquatic models. Mortality rate increased with chain length (PFOA > PFHxA ≫ PFBA) in both whole embryo/larvae and embryonic cell models. The sulfonate group enhanced toxicity with perfluorobutane sulfonate (PFBS) showing higher toxicity than PFBA and PFHxA in both larvae and cells. Toxicity trends were similar among different aquatic models, but sensitivities varied. Discrepancies with other zebrafish studies were confirmed to be associated with a lack of neutralization of acidic pH of dosing solutions in these other investigations, demonstrating the need for rigor in reporting pH of exposure solutions in all experiments. The zebrafish embryonic cell line was also found to be similar to most other cell lines regardless of exposure length. Overall, results agree with findings in other cell lines and organisms where longer chain length and sulfonate group increase toxicity, except in investigations not neutralizing the exposure solutions for these acidic compounds.


Asunto(s)
Caproatos/toxicidad , Caprilatos/toxicidad , Fluorocarburos/toxicidad , Ácidos Sulfónicos/toxicidad , Contaminantes Químicos del Agua/toxicidad , Pez Cebra , Animales , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/embriología , Desarrollo Embrionario/efectos de los fármacos , Pez Cebra/embriología , Pez Cebra/crecimiento & desarrollo
20.
J Mater Chem B ; 8(46): 10609-10615, 2020 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-33136104

RESUMEN

In this work, an anionic conjugated polyelectrolyte (PCP-SO3K), in which the backbone contains alternating 4,4-bis-alkyl-4H-cyclopenta-[2,1-b;3,4-b']-dithiophene and benzene structural units and the charges are provided by pendant sulfonate groups, was synthesized. The ionic nature of PCP-SO3K renders it soluble in water, and PCP-SO3K aqueous solution exhibits good photostability, with two main absorbance bands centered at 490 nm and 837 nm before and after laser irradiation. Its NIR absorption in water, negligible photoluminescence and insignificant intersystem crossing endow PCP-SO3K with efficient photothermal therapy performance, and an effective photothermal conversion efficiency of 56.7% was realized. Thus, PCP-SO3K aqueous solution can be used as an effective photothermal agent for in vivo applications as its photoactivity can be triggered by NIR light and can convert laser energy into thermal energy in a water environment. Of particular importance is the fact that complete tumor remission without recurrence in 4T1 tumor-bearing mice was realized after intravenous injection of PCP-SO3K aqueous solution and laser irradiation (2.0 W cm-2, 808 nm). The results indicate that the application of anionic conjugated polyelectrolytes as photothermal agents in photothermal therapy provides a new platform for the design of photothermal agents for clinical cancer treatment.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Polielectrolitos/uso terapéutico , Ácidos Sulfónicos/uso terapéutico , Tiofenos/uso terapéutico , Animales , Antineoplásicos/efectos de la radiación , Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Femenino , Rayos Infrarrojos , Ratones Endogámicos BALB C , Terapia Fototérmica/métodos , Polielectrolitos/efectos de la radiación , Polielectrolitos/toxicidad , Ácidos Sulfónicos/efectos de la radiación , Ácidos Sulfónicos/toxicidad , Tiofenos/efectos de la radiación , Tiofenos/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...