Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Environ Res ; 212(Pt A): 113157, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35318009

RESUMEN

BACKGROUND: Per- and polyfluoroalkyl substances (PFAS) are a large family of persistent industrial chemicals with endocrine disrupting properties. OBJECTIVES: To examine biomarkers of reproductive function in young adult males according to current environmental exposure to single and combined PFAS. METHODS: The study population consisted of young men (n = 1041, age 18-21) from the Fetal Programming of Semen Quality (FEPOS) cohort. These men were recruited from pregnancies included in the Danish National Birth Cohort (DNBC) between 1996 and 2002. From 2017 to 2019, participants answered an online questionnaire, completed a clinical examination and provided a blood and a semen sample. Exposure to 15 PFAS was measured in plasma. Six compounds were quantified above the limit of detection in at least 80% of the participants. We applied negative binomial regression and weighted quantile sum (WQS) regression models to assess associations between single and combined exposure to PFAS and measures of semen quality, testicular volume and reproductive hormones among the young men. RESULTS: We found no consistent associations between plasma concentrations of PFAS, semen quality and testicular volume. Higher levels of single and combined PFAS were associated with slightly higher levels of follicle-stimulating hormone (FSH) (WQS 4% difference, 95% confidence interval: 0, 9). Perfluorooctanoic acid (PFOA) was the main contributor to this finding with positive signals also from perfluorodecanoic acid (PFDA) and perfluorohexane sulfonic acid (PFHxS). DISCUSSION: We examined exposure to a range of common PFAS in relation to biomarkers of male reproductive function and found an association with higher levels of FSH among young men from the general population in Denmark. Further studies on especially combined exposure to PFAS are needed to expand our understanding of potential endocrine disruption from both legacy and emerging compounds in relation to male reproductive function.


Asunto(s)
Ácidos Alcanesulfónicos , Exposición a Riesgos Ambientales , Contaminantes Ambientales , Fluorocarburos , Genitales Masculinos , Adolescente , Adulto , Ácidos Alcanesulfónicos/administración & dosificación , Estudios Transversales , Exposición a Riesgos Ambientales/efectos adversos , Contaminantes Ambientales/efectos adversos , Fluorocarburos/efectos adversos , Hormona Folículo Estimulante/sangre , Genitales Masculinos/efectos de los fármacos , Humanos , Masculino , Análisis de Semen , Adulto Joven
2.
J Surg Res ; 252: 16-21, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32217350

RESUMEN

BACKGROUND: Tumor-associated glycoprotein (TAG)-72 is a pancarcinoma antigen that is overexpressed in greater than 80% of colorectal adenocarcinomas. CC49 is a TAG-72-specific antibody. The aim of the present study was to demonstrate selective imaging of colon tumors and metastases with the humanized TAG-72 antibody (anti-huCC49) conjugated to a near-infrared fluorophore in orthotopic mouse models. METHODS: Anti-huCC49 was conjugated to near-infrared dye IR800CW. Mouse imaging was performed with the Pearl Trilogy Small Animal and FLARE Imaging Systems. Subcutaneous mouse models of colon cancer cell line LS174T were used to determine the optimal dose of administration and timing of imaging. Orthotopic mouse models of LS174T were established by surgical orthotopic implantation of LS174T tumors onto the serosa of the cecum. Peritoneal carcinomatosis models were established by injection of LS174T cells into the peritoneum of nude mice. Mice were administered anti-huCC49-IR800 via tail vein injection. Mice were euthanized 72 h later and imaged after laparotomy. RESULTS: Subcutaneous LS174T xenografts demonstrated optimal tumor detection 72 h after administration with 50 µg anti-huCC49-IR800CW. Tumors were visualized with fluorescence imaging with a mean tumor-to-liver ratio of 7.39 (standard deviation: 2.76). In the orthotopic model, metastases smaller than 1 mm were fluorescently visualized that were invisible with bright light. CONCLUSIONS: Anti-huCC49-IR800CW provides sensitive and specific imaging of colon cancer and metastases at a submillimeter resolution in metastatic nude mice models. This provides a promising near-infrared probe for the imaging of colon cancer and metastases for preoperative diagnosis and fluorescence-guided surgery.


Asunto(s)
Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Antineoplásicos/administración & dosificación , Antígenos de Neoplasias/inmunología , Neoplasias del Colon/diagnóstico por imagen , Neoplasias Peritoneales/diagnóstico por imagen , Ácidos Alcanesulfónicos/administración & dosificación , Ácidos Alcanesulfónicos/química , Animales , Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Antineoplásicos/química , Anticuerpos Antineoplásicos/inmunología , Línea Celular Tumoral , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Neoplasias del Colon/cirugía , Colorantes Fluorescentes/administración & dosificación , Colorantes Fluorescentes/química , Humanos , Inmunoconjugados/administración & dosificación , Inmunoconjugados/química , Inmunoconjugados/inmunología , Indoles/administración & dosificación , Indoles/química , Ratones , Neoplasias Peritoneales/inmunología , Neoplasias Peritoneales/secundario , Neoplasias Peritoneales/cirugía , Cuidados Preoperatorios/métodos , Espectroscopía Infrarroja Corta/métodos , Cirugía Asistida por Computador/métodos , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Toxicol Appl Pharmacol ; 381: 114715, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31437491

RESUMEN

Exposure of a variety of experimental animals to perfluorooctane sulfonate (PFOS) has shown that it is a potent endocrine-disrupting chemical. However, its interaction with the circadian rhythm on responses along the hypothalamic - pituitary - gonadal - liver (HPGL) axis should be of significant value but has not been adequately investigated. In present study, the effects of PFOS on fecundity, levels of estradiol (E2) and expression of certain genes on the HPGL axis at two time points (8:00 AM and 7:00 PM) were compared after female zebrafish were exposed to 0, 2, 20 and 200 µg/L PFOS for 21 days. In brain, expressions of gonadotropin-releasing hormone (GnRH), gonadotropin-releasing hormone receptor (GnRHr), follicle-stimulating hormone (FSH) and luteinizing hormone (LH) were significantly different after the exposure when sampled at 8:00 AM and at 7:00 PM (P < .05). In liver, significant down-regulation of vitellogenin1 (VTG1) and estrogenic receptor α (ERα) were observed at 7:00 PM compared with 8:00 AM (P < .05). In ovary, the level of CYP19 was significantly different at the two time points (P < .05). The increase of E2 after exposure to 20 µg/L PFOS at 8:00 AM caused compensatory down-regulation of GnRHr and up-regulation of VTG1 and ERα, but not at 7:00 PM. Profiles of concentrations of E2 and several gene expressions alongside the HPGL axis were different between two times points. The change of E2 and gene expressions were more perturbed by PFOS at 8:00 AM than at 7:00 PM with circadian rhythm.


Asunto(s)
Ácidos Alcanesulfónicos/toxicidad , Disruptores Endocrinos/toxicidad , Fertilidad/efectos de los fármacos , Fluorocarburos/toxicidad , Transcriptoma/efectos de los fármacos , Ácidos Alcanesulfónicos/administración & dosificación , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Ritmo Circadiano , Disruptores Endocrinos/administración & dosificación , Estradiol/metabolismo , Femenino , Fluorocarburos/administración & dosificación , Hormona Folículo Estimulante/genética , Hormona Liberadora de Gonadotropina/genética , Hígado/efectos de los fármacos , Hígado/metabolismo , Hormona Luteinizante/genética , Ovario/efectos de los fármacos , Ovario/metabolismo , Receptores LHRH/genética , Pez Cebra
4.
Chem Res Toxicol ; 32(8): 1656-1669, 2019 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-31340646

RESUMEN

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.


Asunto(s)
Ácidos Alcanesulfónicos/farmacología , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Fluorocarburos/farmacología , Metabolómica , Compuestos de Metilmercurio/farmacología , Ácidos Alcanesulfónicos/administración & dosificación , Ácidos Alcanesulfónicos/análisis , Animales , Encéfalo/patología , Relación Dosis-Respuesta a Droga , Femenino , Fluorocarburos/administración & dosificación , Fluorocarburos/análisis , Masculino , Compuestos de Metilmercurio/administración & dosificación , Compuestos de Metilmercurio/análisis , Embarazo , Ratas , Ratas Sprague-Dawley
5.
Int J Toxicol ; 37(5): 383-392, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30134762

RESUMEN

Perfluorooctanesulfonic acid (PFOS) is a persistent organic pollutant with worldwide bioaccumulation due to a very long half-life. Perfluorooctanesulfonic acid exposure results in significant hepatic effects including steatosis, proliferation, hepatomegaly, and in rodents, carcinogenesis. The objective of this study was to determine whether PFOS exposure exacerbates nonalcoholic fatty liver disease and nonalcoholic steatohepatitis pathogenesis. Eight-week-old male C57BL/6 J mice (n = 5 per group) were fed ad libitum normal chow diet (ND) alone, 60% high-fat diet (HFD) alone, ND + PFOS, and HFD + PFOS (0.0001% w/w (1 mg/kg) of PFOS) for 6 weeks. Both HFD alone and the ND + PFOS treatment induced significant adiposity and hepatomegaly, but the HFD + PFOS treatment showed a marked protection. Oil Red O staining and quantitative analysis of hepatic lipid content revealed increased hepatic steatosis in ND + PFOS and in HFD alone fed mice, which was prevented in HFD + PFOS treatment. Further studies revealed that ND + PFOS treatment significantly affected expression of lipid trafficking genes to favor steatosis, but these changes were absent in HFD + PFOS group. Specifically, expression of CD36, the major lipid importer in the cells, and peroxisome proliferator-activated receptor gamma (PPARγ), its major regulator, were induced in HFD + no treatment (NT) and ND + PFOS-fed mice but remained unchanged in HFD + PFOS mice. In conclusion, these data indicate that coadministration of PFOS with HFD mitigates steatosis and hepatomegaly induced by HFD and that by PFOS fed in ND diet via regulation of cellular lipid import machinery. These findings suggest dietary lipid content be considered when performing risk management of PFOS in humans and the elucidation of PFOS-induced hepatotoxicity.


Asunto(s)
Ácidos Alcanesulfónicos/administración & dosificación , Dieta Alta en Grasa , Fluorocarburos/administración & dosificación , Enfermedad del Hígado Graso no Alcohólico/prevención & control , Adiposidad/efectos de los fármacos , Animales , Proliferación Celular/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Hepatomegalia/inducido químicamente , Masculino , Ratones Endogámicos C57BL , Aumento de Peso/efectos de los fármacos
6.
Arch Toxicol ; 91(2): 885-895, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27155986

RESUMEN

Perfluorooctane sulfonate (PFOS) is a persistent environmental contaminant. Although studies have described PFOS-induced neurotoxicity in animal brains and neuronal cells, the molecular mechanisms of PFOS-induced neurotoxicity based on the distribution properties, especially during developmental periods, have not been clarified. To clarify the mechanisms of PFOS-induced neuronal vulnerability during developmental periods, we examined changes in glutamate receptor 2 (GluR2) expression and related neurotoxicity in PFOS-treated primary cortical neurons and neonatal rat brains. Exposure of cortical neurons to 1 µM PFOS for 9 days resulted in decreased α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor subunit GluR2 expression, which subsequently enhanced vulnerability to glutamate by increasing intracellular Ca2+ concentrations. The brain-plasma ratio of PFOS in pups was approximately five times higher than that in dams, although there were no differences in liver-plasma ratio between dams and pups. GluR2 expression in pup cerebral cortex decreased after exposure to 2.0 mg/kg PFOS, and kainic acid induced histopathological abnormalities in PFOS-exposed pups. Our findings suggest that decreased neuronal GluR2 expression is involved in PFOS-induced neurotoxicity, especially during the fetal and neonatal periods.


Asunto(s)
Ácidos Alcanesulfónicos/toxicidad , Fluorocarburos/toxicidad , Neuronas/efectos de los fármacos , Receptores AMPA/metabolismo , Administración Oral , Ácidos Alcanesulfónicos/administración & dosificación , Ácidos Alcanesulfónicos/farmacocinética , Animales , Animales Recién Nacidos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/embriología , Relación Dosis-Respuesta a Droga , Agonistas de Aminoácidos Excitadores/farmacología , Femenino , Fluorocarburos/administración & dosificación , Fluorocarburos/farmacocinética , Ácido Kaínico/farmacología , Neuronas/metabolismo , Neuronas/patología , Embarazo , Ratas Wistar , Receptores AMPA/genética , Distribución Tisular
7.
Biochem Biophys Res Commun ; 477(4): 781-785, 2016 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-27363333

RESUMEN

Perfluorooctane sulfonate (PFOS), a persistent organic pollutant, is blamed to be associated with the incidence of insulin resistance in the general human population. In this study, we found that PFOS inhibited the phosphorylation and activation of protein kinase B (AKT), a key mediator of cellular insulin sensitivity, in human hepatoma HepG2 cells. The mRNA level of the gluconeogenic gene PEPCK, a downstream target gene of AKT, was increased in PFOS-treated cells. Due to stimulated gluconeogenesis, insulin-stimulated glucose uptake was decreased in HepG2 cells. In our previous study, we found that PFOS disturbed autophagy in HepG2 cells. We proposed that PFOS could inhibit the activation of AKT through inhibiting mTORC2, a key regulator of autophagy. In this study, we found that the levels of triglyceride were increased in HepG2 cells. PFOS-induced accumulation of hepatic lipids also contributed to the inhibition of AKT. Eventually, the inhibition of AKT led to insulin resistance in PFOS-treated cells. Our data would provide new mechanistic insights into PFOS-induced hepatic insulin resistance.


Asunto(s)
Ácidos Alcanesulfónicos/administración & dosificación , Fluorocarburos/administración & dosificación , Hepatocitos/metabolismo , Resistencia a la Insulina/fisiología , Insulina/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/fisiología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Glucosa/farmacocinética , Células Hep G2 , Hepatocitos/efectos de los fármacos , Humanos , Transducción de Señal/efectos de los fármacos
8.
BMC Cancer ; 16(1): 942, 2016 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-27927180

RESUMEN

BACKGROUND: Colorectal cancer is the second most common cause of cancer deaths for both men and women, and the third most common cause of cancer in the U.S. Toxicity of current chemotherapeutic agents for colorectal cancer, and emergence of drug resistance underscore the need to develop new, potentially less toxic alternatives. Our recent cross-sectional study in a large Appalachian population, showed a strong, inverse, dose-response association of serum perfluorooctane sulfonate (PFOS) levels to prevalent colorectal cancer, suggesting PFOS may have therapeutic potential in the prevention and/or treatment of colorectal cancer. In these preliminary studies using a mouse model of familial colorectal cancer, the APCmin mouse, and exposures comparable to those reported in human populations, we assess the efficacy of PFOS for reducing tumor burden, and evaluate potential dose-response effects. METHODS: At 5-6 weeks of age, APCmin mice were randomized to receive 0, 20, 250 mg PFOS/kg (females) or 0, 10, 50 and 200 mg PFOS/kg (males) via their drinking water. At 15 weeks of age, gastrointestinal tumors were counted and scored and blood PFOS levels measured. RESULTS: PFOS exposure was associated with a significant, dose-response reduction in total tumor number in both male and female mice. This inverse dose-response effect of PFOS exposure was particularly pronounced for larger tumors (r2 for linear trend = 0.44 for males, p's <0.001). CONCLUSIONS: The current study in a mouse model of familial adenomatous polyposis offers the first experimental evidence that chronic exposure to PFOS in drinking water can reduce formation of gastrointestinal tumors, and that these reductions are both significant and dose-dependent. If confirmed in further studies, these promising findings could lead to new therapeutic strategies for familial colorectal cancer, and suggest that PFOS testing in both preventive and therapeutic models for human colorectal cancer is warranted.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/fisiología , Poliposis Adenomatosa del Colon/tratamiento farmacológico , Ácidos Alcanesulfónicos/administración & dosificación , Modelos Animales de Enfermedad , Fluorocarburos/administración & dosificación , Poliposis Adenomatosa del Colon/genética , Poliposis Adenomatosa del Colon/patología , Administración Oral , Ácidos Alcanesulfónicos/farmacología , Animales , Relación Dosis-Respuesta a Droga , Femenino , Fluorocarburos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL
9.
Drug Chem Toxicol ; 39(1): 97-103, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-25950456

RESUMEN

Perfluorooctane sulfonate (PFOS) is a man-made fluorosurfactant and global pollutant. PFOS a persistent and bioaccumulative compound, and it is widely distributed in humans and wildlife. Therefore, it was added to Annex B of the Stockholm Convention on Persistent Organic Pollutants in May 2009. Curcumin is a natural polyphenolic compound abundant in the rhizome of the perennial herb turmeric. It is commonly used as a dietary spice and coloring agent in cooking and anecdotally as an herb in traditional Asian medicine. In this study, male rats were treated with three different PFOS doses (0.6, 1.25, and 2.5 mg/kg) and one dose of curcumin, from Curcuma longa (80 mg/kg), and combined three doses of PFOS with 80 mg/kg dose of curcumin by gavage for 30 d at 48 h intervals. Here, we investigated the DNA damage via single-cell gel electrophoresis/comet assay and micronucleus test in rat peripheral blood in vivo. It is found that all doses of PFOS increased micronucleus frequency (p < 0.05) and strongly induced DNA damage in peripheral blood in two different parameters; the damaged cell percent and genetically damage index, and curcumin prevented the formation of DNA damage induced by PFOS. Results showed that curcumin inhibited DNA damage including GDI at certain levels at statistical manner, 30.07%, 54.41%, and 36.99% for 0.6 mg/kg, 1.25 mg/kg, and 2.5 mg/kg.


Asunto(s)
Ácidos Alcanesulfónicos/toxicidad , Curcuma/química , Daño del ADN/efectos de los fármacos , Fluorocarburos/toxicidad , Ácidos Alcanesulfónicos/administración & dosificación , Animales , Ensayo Cometa , Curcumina/aislamiento & purificación , Curcumina/farmacología , Relación Dosis-Respuesta a Droga , Fluorocarburos/administración & dosificación , Masculino , Pruebas de Micronúcleos , Ratas , Ratas Wistar
10.
Int J Mol Sci ; 17(9)2016 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-27626407

RESUMEN

Little information exists about the evaluation of potential developmental immunotoxicity induced by perfluorooctane sulfonate (PFOS), a synthetic persistent and increasingly ubiquitous environmental contaminant. To assess potential sex-specific impacts of PFOS on immunological health in the offspring, using male and female C57BL/6 mice, pups were evaluated for developmental immunotoxic effects after maternal oral exposure to PFOS (0.1, 1.0 and 5.0 mg PFOS/kg/day) during Gestational Days 1-17. Spontaneous TH1/TH2-type cytokines, serum levels of testosterone and estradiol were evaluated in F1 pups at four and eight weeks of age. The study showed that male pups were more sensitive to the effects of PFOS than female pups. At eight weeks of age, an imbalance in TH1/TH2-type cytokines with excess TH2 cytokines (IL-4) was found only in male pups. As for hormone levels, PFOS treatment in utero significantly decreased serum testosterone levels and increased estradiol levels only in male pups, and a significant interaction between sex and PFOS was observed for serum testosterone at both four weeks of age (pinteraction = 0.0049) and eight weeks of age (pinteraction = 0.0227) and for estradiol alternation at four weeks of age (pinteraction = 0.0351). In conclusion, testosterone-mediated endocrine function may be partially involved in the TH1/TH2 imbalance induced by PFOS, and these deficits are detectable among both young and adult mice and may affect males more than females.


Asunto(s)
Ácidos Alcanesulfónicos/toxicidad , Citocinas/metabolismo , Estradiol/sangre , Fluorocarburos/toxicidad , Efectos Tardíos de la Exposición Prenatal/inmunología , Testosterona/sangre , Administración Oral , Ácidos Alcanesulfónicos/administración & dosificación , Animales , Peso Corporal/efectos de los fármacos , Femenino , Fluorocarburos/administración & dosificación , Masculino , Exposición Materna , Ratones , Tamaño de los Órganos/efectos de los fármacos , Embarazo , Caracteres Sexuales , Balance Th1 - Th2
11.
Mol Biol Rep ; 41(6): 3755-9, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24535269

RESUMEN

Fluorinated organic compounds, such as perfluorooctane sulfonate, are stable chemicals with a wide range of industrial applications. The potential toxicity of perfluorooctane sulfonate is not well characterized, and even less known are the mechanisms underlying its toxic effects. Perfluorooctane sulfonate change of inner mitochondrial membrane permeability has been implicated as a potential mechanism of toxicity. In this study, we research that perfluorooctane sulfonate effects the expression of Apaf1 and Caspase3 genes in the amnion and fetal lung cell line that initiate the cells to undergo apoptosis. The expression of Caspase3 and Apaf1 was determined by using quantitative RT-PCR. In the study there is significant increase in expression of Caspase3 and Apaf1 in amnion and fetal lung cell line exposed to high dose (p < 0.001, p = 0.004). Also there is significant increase in cell lines exposed for a long period of time to perfluorooctane sulfonate (p = 0.001). But no significant increase was seen in the low doses and exposed for a short period of time. In conclusion, apoptotic gene expression is increase in cells exposed perfluorooctane sulfonate by dose dependent manner was determined. So this work is the first study examines the apoptotic effects of perfluorooctane sulfonate in human embryonic cells it will lead the way to the other topical studies.


Asunto(s)
Ácidos Alcanesulfónicos/administración & dosificación , Apoptosis/efectos de los fármacos , Fluorocarburos/administración & dosificación , Regulación de la Expresión Génica/efectos de los fármacos , Ácidos Alcanesulfónicos/toxicidad , Factor Apoptótico 1 Activador de Proteasas/biosíntesis , Caspasa 3/biosíntesis , Línea Celular , Permeabilidad de la Membrana Celular/efectos de los fármacos , Células Madre Embrionarias/efectos de los fármacos , Contaminantes Ambientales/administración & dosificación , Contaminantes Ambientales/toxicidad , Fluorocarburos/toxicidad , Humanos , Técnicas In Vitro , Membranas Mitocondriales/efectos de los fármacos
12.
J Immunotoxicol ; 21(1): 2371868, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39066581

RESUMEN

Per- and polyfluoroalkyl substances (PFAS) are a large group of synthetic surfactants of over 12,000 compounds that are incorporated into numerous products for their chemical and physical properties. Studies have associated PFAS with adverse health effects. Although there is a high potential for dermal exposure, these studies are lacking. The present study evaluated the systemic and immunotoxicity of subchronic 28- or 10-days of dermal exposure, respectively, to PFHpS (0.3125-2.5% or 7.82-62.5 mg/kg/dose) or PFOS (0.5% or 12.5 mg/kg/dose) in a murine model. Elevated levels of PFHpS were detected in the serum and urine, suggesting that absorption is occurring through the dermal route. PFHpS induced significantly increased relative liver weight, significantly decreased relative spleen and thymus weight, altered serum chemistries, and altered histopathology. Additionally, PFHpS 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 of genes involved in fatty acid metabolism, necrosis, and inflammation. Immune-cell phenotyping identified significant decreases in B-cells and CD11b+ monocyte and/or macrophages in the spleen along with decreases in eosinophils and dendritic cells in the skin. These findings support PFHpS absorption through the skin leading to liver damage and immune suppression.


Asunto(s)
Ácidos Alcanesulfónicos , Fluorocarburos , Animales , Fluorocarburos/toxicidad , Fluorocarburos/administración & dosificación , Ratones , Ácidos Alcanesulfónicos/toxicidad , Ácidos Alcanesulfónicos/administración & dosificación , Femenino , Humanos , Bazo/efectos de los fármacos , Bazo/inmunología , Bazo/patología , Ácidos Sulfónicos/toxicidad , Ácidos Sulfónicos/administración & dosificación , Administración Tópica , Hígado/efectos de los fármacos , Hígado/inmunología , Hígado/patología , Masculino , Piel/efectos de los fármacos , Piel/inmunología , Piel/patología
13.
Ecotoxicology ; 22(7): 1133-44, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23907449

RESUMEN

Perfluorobutanesulfonate (PFBS), as a substitute for perfluorooctanesulfonate (PFOS), is widespread in the environment and biotic samples as well as PFOS. To investigate effects of PFOS and PFBS on the growth and sexual development of amphibians, we exposed Xenopus laevis tadpoles at a series of concentrations of PFOS and PFBS (0.1; 1; 100; 1,000 µg/l) as well as 17-beta-estradiol (E2, 100 ng/l) and 5 alpha-androstan-17-beta-ol-3-one (DHT, 100 ng/l) from stage 46/47 to 2 months postmetamorphosis. We found that neither PFOS nor PFBS had a significant effect on the survival and growth. However, they caused hepatohistological impairment at higher concentrations (100; 1,000 µg/l). Unlike E2, PFOS at all concentrations did not alter the sex ratio and induce intersex, but caused degeneration of spermatogonia in testes except for the lowest concentration. PFBS had no effect on the sex ratio and gonadal histology. PFOS and PFBS promoted expression of estrogen receptor (ER) and androgen receptor (AR), but not affected aromatase expression in the brain. The increase in expression of ER and AR suggests an increase in the responsiveness to the corresponding sex hormone and potential effects on sexual development. Our results show that PFBS as well as PFOS have adverse effects on hepato-histology and sexual development on X. laevis. Also, PFOS- and PFBS-induced increase in ER and AR expression highlights the need to further study effects of PFOS and PFBS on subsequently gonadal development, sexual dimorphism, and secondary sex characteristics in X. laevis. It is debatable that PFBS is widely used as a substitute of PFOS.


Asunto(s)
Ácidos Alcanesulfónicos/toxicidad , Fluorocarburos/toxicidad , Desarrollo Sexual/efectos de los fármacos , Ácidos Sulfónicos/toxicidad , Contaminantes Químicos del Agua/toxicidad , Xenopus laevis/crecimiento & desarrollo , Ácidos Alcanesulfónicos/administración & dosificación , Animales , Aromatasa/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Estradiol/toxicidad , Fluorocarburos/administración & dosificación , Hormonas Esteroides Gonadales/metabolismo , Gónadas/efectos de los fármacos , Gónadas/crecimiento & desarrollo , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ácidos Sulfónicos/administración & dosificación , Testículo/efectos de los fármacos , Testículo/crecimiento & desarrollo
14.
Biol Reprod ; 84(5): 1016-23, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21209418

RESUMEN

Perfluorooctanesulfonate (PFOS) was produced and used by various industries and in consumer products. Because of its persistence, it is ubiquitous in air, water, soil, wildlife, and humans. Although the adverse effects of PFOS on male fertility have been reported, the underlying mechanisms have not yet been elucidated. Here, for the first time, the effects of PFOS on testicular signaling, such as gonadotropin, growth hormone, insulin-like growth factor, and inhibins/activins were shown to be directly related to male subfertility. Sexually mature 8-wk-old CD1 male mice were administered by gavages in corn oil daily with 0, 1, 5, or 10 mg/kg PFOS for 7, 14, or 21 days. Serum concentrations of testosterone and epididymal sperm counts were significantly lower in the mice after 21 days of the exposure to the highest dose compared with the controls. The expression levels of testicular receptors for gonadotropin, growth hormone, and insulin-like growth factor 1 were considerably reduced on Day 21 in mice exposed daily to 10 or 5 mg/kg PFOS. The transcript levels of the subunits of the testicular factors (i.e., inhibins and activins), Inha, Inhba, and Inhbb, were significantly lower on Day 21 of daily exposure to 10, 5, or 1 mg/kg PFOS. The mRNA expression levels of steroidogenic enzymes (i.e., StAR, CYP11A1, CYP17A1, 3beta-HSD, and 17beta-HSD) were notably reduced. Therefore, PFOS-elicited subfertility in male mice is manifested as progressive deterioration of testicular signaling.


Asunto(s)
Ácidos Alcanesulfónicos/toxicidad , Contaminantes Ambientales/toxicidad , Fluorocarburos/toxicidad , Sistema Hipotálamo-Hipofisario/fisiología , Infertilidad Masculina/inducido químicamente , Transducción de Señal/efectos de los fármacos , Testículo/fisiología , Activinas/genética , Activinas/metabolismo , Ácidos Alcanesulfónicos/administración & dosificación , Animales , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Contaminantes Ambientales/administración & dosificación , Epidídimo/citología , Epidídimo/efectos de los fármacos , Fluorocarburos/administración & dosificación , Infertilidad Masculina/metabolismo , Inhibinas/genética , Inhibinas/metabolismo , Masculino , Ratones , Ratones Endogámicos , Oligospermia/inducido químicamente , ARN Mensajero/metabolismo , Distribución Aleatoria , Receptor IGF Tipo 1/metabolismo , Receptores de Gonadotropina/metabolismo , Receptores de Somatotropina/metabolismo , Testosterona/sangre
15.
Arch Toxicol ; 85(6): 613-21, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21046362

RESUMEN

The toxicity of perfluorooctane sulfonate (PFOS), a persistent organic compound, is of great concern. Several studies have reported that PFOS decreases circulating thyroid hormone (TH) concentrations. However, the mechanisms involved remain to be determined. Female rats were exposed to (1) vehicle; (2) PFOS (0.2, 1.0, and 3.0 mg/kg); (3) propylthiouracil (PTU, 10 mg/kg); or (4) PTU (10 mg/kg) + PFOS (3.0 mg/kg) by gavage once a day for 5 consecutive days. Parameters including contents of total T4 (TT4) and total T3 (TT3) in both serum and bile, serum concentrations of transthyretin and thyroglobulin, as well as transcripts of transporters involved in hepatic uptake and efflux of T4 were determined in control and PFOS-exposed groups. TT4 and TT3 were also analyzed in PTU and PTU + PFOS groups in order to reflect the different hormone effects between PFOS, PTU, and PFOS + PTU. Results showed that serum TT4 and TT3 decreased, while bile TT4 and TT3 remained stable following PFOS exposure. Exposure to 3.0 mg/kg of PFOS significantly enhanced hepatic organic anion transporter OATP2 mRNA expression (1.43 times of control). Treatment with PFOS increased hepatic expression of multidrug resistance--associated protein MRP2, approximately 1.80 and 1.69 times of control in 1.0 and 3.0 mg/kg groups, respectively. Spearman's correlation coefficients revealed that MRP2 mRNA expression correlated well with serum TT4 level (r = -0.528, P = 0.012). Serum thyroglobulin and transthyretin levels remained stable. Serum TT3, bile TT4, and bile TT3 were significantly different between PFOS and PTU groups. No significant differences of TT4 and TT3 in both serum and bile were observed between PTU and PTU + PFOS (P > 0.05). In conclusion, PFOS increased hepatic expression of OAPT2, which could possibly enhance hepatic uptake and metabolism of T4 in rats. PFOS-induced TT4 deficiency is mainly due to the extrathyroidal metabolism of T4, which is probably different from the classic goitrogen, PTU.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Ácidos Alcanesulfónicos/toxicidad , Antitiroideos/toxicidad , Contaminantes Ambientales/toxicidad , Fluorocarburos/toxicidad , Hígado/efectos de los fármacos , Transportadores de Anión Orgánico/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Transportadoras de Casetes de Unión a ATP/genética , Ácidos Alcanesulfónicos/administración & dosificación , Animales , Antitiroideos/administración & dosificación , Bilis/metabolismo , Relación Dosis-Respuesta a Droga , Contaminantes Ambientales/administración & dosificación , Femenino , Fluorocarburos/administración & dosificación , Hipotiroidismo/inducido químicamente , Hígado/metabolismo , Transportadores de Anión Orgánico/genética , Propiltiouracilo/toxicidad , ARN Mensajero/metabolismo , Distribución Aleatoria , Ratas , Ratas Wistar , Tiroxina/sangre , Tiroxina/metabolismo , Triyodotironina/sangre , Triyodotironina/metabolismo
16.
Pharmazie ; 66(5): 357-61, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21699069

RESUMEN

PX-18 and PX-13 are secretory phospholipase A2-IIA (sPLA2-IIA) inhibitors. An increased expression of sPLA2 in psoriatic skin has been reported. The selective inhibition of this enzyme is a new therapeutic approach. For dermal application PX-18 and PX-13 have been loaded to Nanostructured lipid carriers (NLC). The PX-18-loaded and PX-13-loaded NLC possessed an average particles size of about 250 nm, a narrow particle size distribution (PI < 0.2), a high entrapment efficiency as well as a good physical stability, as already indicated by their high zeta potential. Both NLC formulations have been incorporated into a hydroxyethyl cellulose gel and an o/w cream. In the gel and in the o/w cream PX-18-loaded and PX-13-loaded NLC showed a good physical stability. Neither aggregation nor dissolution of NLC took place.


Asunto(s)
Alcanosulfonatos/administración & dosificación , Ácidos Alcanesulfónicos/administración & dosificación , Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos , Liposomas/química , Nanoestructuras/química , Ácidos Oléicos/administración & dosificación , Inhibidores de Fosfolipasa A2 , Administración Tópica , Alcanosulfonatos/química , Ácidos Alcanesulfónicos/química , Rastreo Diferencial de Calorimetría , Celulosa/análogos & derivados , Química Farmacéutica , Electroquímica , Emulsiones , Rayos Láser , Ácidos Oléicos/química , Tamaño de la Partícula , Fotones , Absorción Cutánea , Espectrofotometría , Termodinámica
17.
Toxicology ; 464: 152997, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34695511

RESUMEN

Per- and polyfluoroalkyl substances (PFAS) represent a large class of structurally diverse chemicals of increasing public concern, mostly due to their chemical stability and undetermined toxicity profiles. In laboratory animals, adverse effects implicated for certain PFAS, perfluorooctanoic acid (PFOA) and perfluorooctane sulfonate (PFOS) in particular, include liver toxicity and the associated metabolic dysregulation, immune and thyroid alterations, reproductive toxicity, and selected tumors. The broad commercialization and environmental distribution of PFAS has drawn attention to the need for understanding risks associated with combined exposure to multiple PFAS in complex mixtures. The purpose of this investigation is to determine whether binary combinations of PFAS elicit a molecular response that is either greater than or less than the sum of the individual responses. Exposure of FaO rat hepatoma cells for 24 h to 25 µM-200 µM of the 4- and 8-carbon perfluorocarboxylic acids (PFBA and PFOA) or the 4, 6, and 8-carbon perfluorosulfonic acids (PFBS, PFHxS, and PFOS, respectively) individually caused a dose-dependent increase in PPARα-regulated expression of peroxisomal bifunctional enzyme (Ehhadh). Potency increased with carbon number, with the carboxylates eliciting a greater transcriptional response than the corresponding sulfonates. Combined exposure to PFOA and PFBA produced an effect that was significantly less than the sum of the individual responses. The response to the combination of PFOA and PFOS produced a summative effect at concentrations that were not cytotoxic. Combined exposures to PFOS and either PFBS or PFHxS at low noncytotoxic concentrations produced a transcriptional effect that was significantly less than the sum of the individual effects. The results demonstrate that among the five structurally related perfluoroalkyl acids included in this investigation, PPARα transcriptional activation in response to combined binary exposures is consistently at or below that predicted by the sum of the individual effects.


Asunto(s)
Ácidos Alcanesulfónicos/toxicidad , Caprilatos/toxicidad , Fluorocarburos/toxicidad , PPAR alfa/metabolismo , Ácidos Alcanesulfónicos/administración & dosificación , Ácidos Alcanesulfónicos/química , Animales , Caprilatos/administración & dosificación , Caprilatos/química , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Contaminantes Ambientales/administración & dosificación , Contaminantes Ambientales/química , Contaminantes Ambientales/toxicidad , Fluorocarburos/administración & dosificación , Fluorocarburos/química , Humanos , Neoplasias Hepáticas/metabolismo , Ratas
18.
Toxicology ; 464: 153028, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34762985

RESUMEN

Perfluorooctane sulfonate (PFOS) has been added to Stockholm Convention for global phase out, but will continue to contribute to the chemical burden in humans for a long time to come due to extreme persistence in the environment. In the body, PFOS is transferred into to the ovarian follicular fluid that surrounds the maturing oocyte. In the present study, bovine cumulus oocyte complexes were exposed to PFOS during 22 h in vitro maturation. Concentrations of 2 ng g-1 (PFOS-02) representing average human exposure and 53 ng g-1 (PFOS-53) relevant to highly exposed groups were used. After exposure, developmental competence was recorded until day 8 after fertilisation. Blastocysts were fixed and either stained to evaluate blastomere number and lipid distribution using confocal microscopy or frozen and pooled for microarray-based gene expression and DNA methylation analyses. PFOS-53 delayed the first cleavage to two-cell stage and beyond at 44 h after fertilisation (p < .01). No reduction of proportion blastocysts were seen at day 8 in either of the groups, but PFOS-53 exposure resulted in delayed development into more advanced stages of blastocysts seen as both reduced developmental stage (p = .001) and reduced number of blastomeres (p = .04). Blastocysts showed an altered lipid distribution that was more pronounced after exposure to PFOS-53 (increased total lipid volume, p=.0003, lipid volume/cell p < .0001) than PFOS-02, where only decreased average lipid droplet size (p=.02) was observed. Gene expression analyses revealed pathways differently regulated in the PFOS-treated groups compared to the controls, which were related to cell death and survival through e.g., P38 mitogen-activated protein kinases and signal transducer and activator of transcription 3, which in turn activates tumour protein 53 (TP53). Transcriptomic changes were also associated with metabolic stress response, differentiation and proliferation, which could help to explain the phenotypic changes seen in the blastocysts. The gene expression changes were more pronounced after exposure to PFOS-53 compared to PFOS-02. DNA-methylation changes were associated with similar biological functions as the transcriptomic data, with the most significantly associated pathway being TP53. Collectively, these results reveal that brief PFOS exposure during oocyte maturation alters the early embryo development at concentrations relevant to humans. This study adds to the evidence that PFOS has the potential to affect female fertility.


Asunto(s)
Ácidos Alcanesulfónicos/toxicidad , Desarrollo Embrionario/efectos de los fármacos , Fluorocarburos/toxicidad , Oocitos/efectos de los fármacos , Ácidos Alcanesulfónicos/administración & dosificación , Animales , Blastocisto/citología , Blastocisto/efectos de los fármacos , Bovinos , Metilación de ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Fluorocarburos/administración & dosificación , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Microscopía Confocal
19.
J Toxicol Sci ; 45(12): 763-767, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33268676

RESUMEN

A simplified physiologically based pharmacokinetic (PBPK) model consisting of chemical receptor, metabolizing and/or excreting, and central compartments was recently proposed. In the current study, this type of PBPK model was set up for perfluorooctane sulfonate, an environmental toxicant with liver effects, as a model compound; the model was then used to estimate tissue concentrations. The pharmacokinetic parameter input values for the model were calculated to give the best fit to reported/measured blood substrate concentrations in rats. The maximum concentrations and areas under the concentration versus time curves in plasma, liver, and kidney extrapolated using PBPK models for perfluorobutane sulfonic acid, perfluorohexane sulfonic acid, and perfluorooctane sulfonic acid were consistent with the reported mean values in rats. Using the rat models and scaled-up human PBPK models, some accumulation of perfluorooctane sulfonic acid in plasma and liver was seen after repeated doses. The reported 50th and 95th percentile concentrations of perfluorooctane sulfonic acid in human blood (0.0048 and 0.0183 ng/mL, respectively) in the general population underwent reverse dosimetry analysis using our PBPK models. These human blood concentrations potentially imply exposures of 0.041 and 0.16 µg/kg/day, respectively, for 90 days, values that are roughly similar to the reference dose (0.02 µg/kg/day) with an uncertainty factor of 30. These results indicate the relatively good estimates for tissue and blood exposures of chemical substrates after oral doses generated using the latest PBPK models.


Asunto(s)
Ácidos Alcanesulfónicos/farmacocinética , Ácidos Alcanesulfónicos/toxicidad , Fluorocarburos/farmacocinética , Fluorocarburos/toxicidad , Riñón/metabolismo , Hígado/metabolismo , Modelos Biológicos , Administración Oral , Ácidos Alcanesulfónicos/administración & dosificación , Ácidos Alcanesulfónicos/sangre , Animales , Relación Dosis-Respuesta a Droga , Fluorocarburos/administración & dosificación , Fluorocarburos/sangre , Humanos , Nivel sin Efectos Adversos Observados , Ratas , Distribución Tisular , Toxicocinética
20.
Toxicology ; 445: 152610, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33027616

RESUMEN

Perfluoroalkyl acids (PFAAs) are persistent man-made chemicals, ubiquitous in nature and present in human samples. Although restrictions are being introduced, they are still used in industrial processes as well as in consumer products. PFAAs cross the blood-brain-barrier and have been observed to induce adverse neurobehavioural effects in humans and animals as well as adverse effects in neuronal in vitro studies. The sulfonated PFAA perfluorooctane sulfonic acid (PFOS), has been shown to induce excitotoxicity via the N-methyl-D-aspartate receptor (NMDA-R) in cultures of rat cerebellar granule neurons (CGNs). In the present study the aim was to further characterise PFOS-induced toxicity (1-60 µM) in rat CGNs, by examining interactions between PFOS and elements of glutamatergic signalling and excitotoxicity. Effects of the carboxylated PFAA, perfluorooctanoic acid (PFOA, 300-500 µM) on the same endpoints were also examined. During experiments in immature cultures at days in vitro (DIV) 8, PFOS increased both the potency and efficacy of glutamate, whereas in mature cultures at DIV 14 only increased potency was observed. PFOA also increased potency at DIV 14. PFOS-enhanced glutamate toxicity was further antagonised by the competitive NMDA-R antagonist 3-((R)-2-Carboxypiperazin-4-yl)-propyl-1-phosphonic acid (CPP) at DIV 8. At DIV 8, PFOS also induced glutamate release (9-13 fold increase vs DMSO control) after 1-3 and 24 h exposure, whereas for PFOA a large (80 fold) increase was observed, but only after 24 h. PFOS and PFOA both also increased alanine and decreased serine levels after 24 h exposure. In conclusion, our results indicate that PFOS at concentrations relevant in an occupational setting, may be inducing excitotoxicity, and potentiation of glutamate signalling, via an allosteric action on the NMDA-R or by actions on other elements regulating glutamate release or NMDA-R function. Our results further support our previous findings that PFOS and PFOA at equipotent concentrations induce toxicity via different mechanisms of action.


Asunto(s)
Ácidos Alcanesulfónicos/toxicidad , Caprilatos/toxicidad , Cerebelo/efectos de los fármacos , Agonistas de Aminoácidos Excitadores/toxicidad , Fluorocarburos/toxicidad , Ácido Glutámico/toxicidad , Neuronas/efectos de los fármacos , Ácidos Alcanesulfónicos/administración & dosificación , Animales , Caprilatos/administración & dosificación , Bovinos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Cerebelo/patología , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Agonistas de Aminoácidos Excitadores/administración & dosificación , Femenino , Fluorocarburos/administración & dosificación , Ácido Glutámico/administración & dosificación , Masculino , Neuronas/patología , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA