Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Cell Biochem Funct ; 42(4): e4060, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38816947

RESUMO

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


Assuntos
Ácidos Alcanossulfônicos , Apoptose , Fluorocarbonos , Proteína 1 Associada a ECH Semelhante a Kelch , Fator 2 Relacionado a NF-E2 , Animais , Ratos , Ácidos Alcanossulfônicos/farmacologia , Ácidos Alcanossulfônicos/toxicidade , Apoptose/efeitos dos fármacos , Fator 2 Relacionado a NF-E2/metabolismo , Fluorocarbonos/farmacologia , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Masculino , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Inflamação/induzido quimicamente , Inflamação/patologia , Flavonas/farmacologia
2.
Biomed Res Int ; 2021: 6625952, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33880372

RESUMO

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


Assuntos
Ácidos Alcanossulfônicos/farmacologia , Apoptose , Autofagia , Embrião de Mamíferos/patologia , Fluorocarbonos/farmacologia , Fígado/embriologia , Fígado/patologia , Espécies Reativas de Oxigênio/metabolismo , Acetilcisteína/farmacologia , Adenina/análogos & derivados , Adenina/farmacologia , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Cadaverina/farmacologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Microambiente Celular/efeitos dos fármacos , Humanos , Fígado/efeitos dos fármacos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Proteínas/metabolismo , Vacúolos/efeitos dos fármacos , Vacúolos/metabolismo
3.
Immunol Lett ; 233: 31-41, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33722553

RESUMO

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


Assuntos
Ácidos Alcanossulfônicos/farmacologia , Diferenciação Celular/efeitos dos fármacos , Fluorocarbonos/farmacologia , Granulócitos/efeitos dos fármacos , Granulócitos/imunologia , Fatores Imunológicos/farmacologia , Linfócitos/efeitos dos fármacos , Linfócitos/imunologia , Animais , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Biomarcadores , Medula Óssea/efeitos dos fármacos , Medula Óssea/imunologia , Medula Óssea/metabolismo , Diferenciação Celular/imunologia , Feminino , Granulócitos/citologia , Granulócitos/metabolismo , Imunofenotipagem , Vírus da Influenza A/imunologia , Linfócitos/citologia , Linfócitos/metabolismo , Masculino , Camundongos , Especificidade de Órgãos , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/metabolismo , Infecções por Orthomyxoviridae/virologia , Baço/citologia , Baço/efeitos dos fármacos , Baço/imunologia , Baço/metabolismo , Timo/imunologia , Timo/metabolismo
4.
Chem Res Toxicol ; 33(6): 1478-1486, 2020 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-32423201

RESUMO

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


Assuntos
Ácidos Alcanossulfônicos/farmacologia , Éteres/farmacologia , Fluorocarbonos/farmacologia , Albumina Sérica Humana/metabolismo , Linhagem Celular Tumoral , Humanos , Simulação de Acoplamento Molecular
5.
Biomed Res Int ; 2020: 8818160, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33457418

RESUMO

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


Assuntos
Ácidos Alcanossulfônicos/farmacologia , Fígado Gorduroso/tratamento farmacológico , Fluorocarbonos/farmacologia , Proantocianidinas/farmacologia , Animais , Antígenos CD36/metabolismo , Colesterol/metabolismo , Proteínas de Ligação a Ácido Graxo/metabolismo , Peróxido de Hidrogênio , Inflamação , Interleucina-6/metabolismo , Metabolismo dos Lipídeos/efeitos dos fármacos , Peroxidação de Lipídeos , Fígado/efeitos dos fármacos , Masculino , Malondialdeído/metabolismo , Camundongos , Estresse Oxidativo , Triglicerídeos/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
6.
Chemosphere ; 237: 124402, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31352096

RESUMO

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


Assuntos
Ácidos Alcanossulfônicos/farmacologia , Fluorocarbonos/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Alcanossulfonatos/farmacologia , Caprilatos/farmacologia , Diferenciação Celular/efeitos dos fármacos , Éter , Perfil Genético , Humanos , Células-Tronco Mesenquimais/citologia , Osteogênese/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
7.
Chemosphere ; 200: 283-294, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29494909

RESUMO

Perfluorooctane sulfonate (PFOS), one member of polyfluoroalkyl chemicals (PFASs), persist in the environment and are found in relatively high concentrations in animal livers. PFOS has been shown to induce tumour of the liver in rats following chronic dietary administration. However, the molecular mechanisms involved in PFOS-induced hepatocellular hypertrophy are still not well characterized. In this study, male Sprague-Dawley rats were daily gavaged with PFOS (1 or 10 mg/kg body weight) for 28 days. Rat primary cultured Kupffer cells or hepatocytes were exposed to 100 µM PFOS for 0-48 h. Our results showed that PFOS exposure caused serious hepatocellular damage and obvious inflammatory cell infiltration and increased serum tumour necrosis factor-ɑ (TNF-α) and interleukin-6 (IL-6) levels. Particularly, PFOS exposure triggered Kupffer cell activation and significantly upregulated the expression of proliferating cell nuclear antigen (PCNA), c-Jun, c-MYC and Cyclin D1 (CyD1) in liver. In vitro, PFOS significantly induced production of TNF-α and IL-6 in Kupffer cells and increased PCNA, c-Jun, c-MYC and CyD1 expression in the primary hepatocytes co-cultured with Kupffer cells. However, Kupffer cell activation was mostly abolished by anti-TNF-α or anti-IL6 treatment. Furthermore, blockage of TNF-α and IL-6 significantly inhibited hepatocyte proliferation by gadolinium chloride (GdCl3) pre-treatment in PFOS-treated mice and primary cultured Kupffer cells. On the other hand, NF-κB inhibitor (PDTC) and c-Jun amino-terminal kinase (JNK) inhibitor (SP600125) significantly inhibited production of PFOS-induced TNF-α and IL-6. Taken together, these data suggest that PFOS induces Kupffer cell activation, leading to hepatocyte proliferation by through the NF-κB/TNF-ɑ/IL-6-dependent pathway.


Assuntos
Ácidos Alcanossulfônicos/farmacologia , Proliferação de Células/efeitos dos fármacos , Fluorocarbonos/farmacologia , Hepatócitos/citologia , Interleucina-6/metabolismo , Células de Kupffer/fisiologia , NF-kappa B/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Células Cultivadas , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Interleucina-6/genética , Células de Kupffer/citologia , Células de Kupffer/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/genética , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/genética
8.
Chemosphere ; 191: 1056-1064, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28939271

RESUMO

Perfluorooctane sulphonate (PFOS) has been reported to accumulate in liver and cause damage. The molecular mechanism of the PFOS-induced hepatotoxicity has not been completely elucidated. The aim of the present study was to investigate whether PFOS-induced oxidative stress plays an important role in liver damage, and if so, what pathway it undergoes for the mechanism of its toxicological action. Male Sprague-Dawley (SD) rats were orally administrated with PFOS at single dose of 1 or 10 mg/kg body weight for 28 consecutive days. Increased serum levels of liver enzymes and abnormal ultra structural changes were observed in the PFOS-exposed rats. Particularly, PFOS exposure significantly increased intracellular reactive oxygen species (ROS) and nitric oxide (NO) production, but weakened intracellular antioxidant defence by inhibiting catalase and superoxide dismutase activities. Signal transduction studies showed that PFOS exposure significantly elevated inducible nitric oxide synthase (iNOS), Bax, cytochrome c, cleaved caspase-9 and cleaved caspase-3, indicating the mitochondria-dependent apoptotic pathway was activated. On the other hand, significant alterations of the PFOS-induced protein expression of NF-κB and IκBα in association with an enhanced level of TNF-α were observed. Taken together, these results indicate that mitochondria play an important role in PFOS-induced hepatotoxicity.


Assuntos
Ácidos Alcanossulfônicos/farmacologia , Fluorocarbonos/farmacologia , Fígado/efeitos dos fármacos , Mitocôndrias/metabolismo , NF-kappa B/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo , Ácidos Alcanossulfônicos/administração & dosagem , Ácidos Alcanossulfônicos/toxicidade , Animais , Antioxidantes/metabolismo , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Caspase 9/metabolismo , Fluorocarbonos/administração & dosagem , Fluorocarbonos/toxicidade , Fígado/lesões , Fígado/metabolismo , Fígado/patologia , Masculino , Óxido Nítrico Sintase Tipo II/metabolismo , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos
9.
Ann Vasc Surg ; 42: 293-298, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28300679

RESUMO

BACKGROUND: Arterial blood pressure-induced shear stress causes endothelial cell apoptosis and inflammation in vein grafts after coronary artery bypass grafting. As the inflammatory protein type IIA secretory phospholipase A2 (sPLA2-IIA) has been shown to progress atherosclerosis, we hypothesized a role for sPLA2-IIA herein. METHODS: The effects of PX-18, an inhibitor of both sPLA2-IIA and apoptosis, on residual endothelium and the presence of sPLA2-IIA were studied in human saphenous vein segments (n = 6) perfused at arterial blood pressure with autologous blood for 6 hrs. RESULTS: The presence of PX-18 in the perfusion blood induced a significant 20% reduction in endothelial cell loss compared to veins perfused without PX18, coinciding with significantly reduced sPLA2-IIA levels in the media of the vein graft wall. In addition, PX-18 significantly attenuated caspase-3 activation in human umbilical vein endothelial cells subjected to shear stress via mechanical stretch independent of sPLA2-IIA. CONCLUSIONS: In conclusion, PX-18 protects saphenous vein endothelial cells from arterial blood pressure-induced death, possibly also independent of sPLA2-IIA inhibition.


Assuntos
Ácidos Alcanossulfônicos/farmacologia , Pressão Arterial , Células Endoteliais/efeitos dos fármacos , Fosfolipases A2 do Grupo II/antagonistas & inibidores , Mecanotransdução Celular/efeitos dos fármacos , Ácidos Oleicos/farmacologia , Inibidores de Fosfolipase A2/farmacologia , Veia Safena/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Células Cultivadas , Células Endoteliais/enzimologia , Células Endoteliais/patologia , Fosfolipases A2 do Grupo II/metabolismo , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/enzimologia , Células Endoteliais da Veia Umbilical Humana/patologia , Humanos , Veia Safena/enzimologia , Veia Safena/patologia , Fatores de Tempo
10.
J Cereb Blood Flow Metab ; 37(4): 1199-1212, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27193034

RESUMO

Lipid sensor peroxisome proliferator-activated receptor alpha (PPAR- α) is the master regulator of lipid metabolism. Dietary release of endogenous free fatty acids, fibrates, and certain persistent environmental pollutants, e.g. perfluoroalkyl fire-fighting foam components, are peroxisome proliferator-activated receptor alpha ligands. Here, we define a role for peroxisome proliferator-activated receptor alpha in regulating the expression of three ATP-driven drug efflux transporters at the rat and mouse blood-brain barriers: P-glycoprotein (Abcb1), breast cancer resistance protein (Bcrp/Abcg2), and multidrug resistance-associated protein 2 (Mrp2/Abcc2). Exposing isolated rat brain capillaries to linoleic acid, clofibrate, or PKAs increased the transport activity and protein expression of the three ABC transporters. These effects were blocked by the PPAR- α antagonist, GW6471. Dosing rats with 20 mg/kg or 200 mg/kg of clofibrate decreased the brain accumulation of the P-glycoprotein substrate, verapamil, by 50% (in situ brain perfusion; effects blocked by GW6471) and increased P-glycoprotein expression and activity in capillaries ex vivo. Fasting C57Bl/6 wild-type mice for 24 h increased both serum lipids and brain capillary P-glycoprotein transport activity. Fasting did not alter P-glycoprotein activity in PPAR- α knockout mice. These results indicate that hyperlipidemia, lipid-lowering fibrates and exposure to certain fire-fighting foam components activate blood-brain barrier peroxisome proliferator-activated receptor alpha, increase drug efflux transporter expression and reduce drug delivery to the brain.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Barreira Hematoencefálica/metabolismo , Regulação da Expressão Gênica , PPAR alfa/fisiologia , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/genética , Ácidos Alcanossulfônicos/farmacologia , Animais , Transporte Biológico , Barreira Hematoencefálica/efeitos dos fármacos , Encéfalo/irrigação sanguínea , Capilares/efeitos dos fármacos , Capilares/metabolismo , Clofibrato/farmacologia , Jejum/metabolismo , Fluorocarbonos/farmacologia , Ácido Linoleico/farmacologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 2 Associada à Farmacorresistência Múltipla , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Oxazóis/farmacologia , PPAR alfa/agonistas , PPAR alfa/antagonistas & inibidores , PPAR alfa/genética , Ratos Sprague-Dawley , Tirosina/análogos & derivados , Tirosina/farmacologia
11.
BMC Cancer ; 16(1): 942, 2016 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-27927180

RESUMO

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.


Assuntos
Proteína da Polipose Adenomatosa do Colo/fisiologia , Polipose Adenomatosa do Colo/tratamento farmacológico , Ácidos Alcanossulfônicos/administração & dosagem , Modelos Animais de Doenças , Fluorocarbonos/administração & dosagem , Polipose Adenomatosa do Colo/genética , Polipose Adenomatosa do Colo/patologia , Administração Oral , Ácidos Alcanossulfônicos/farmacologia , Animais , Relação Dose-Resposta a Droga , Feminino , Fluorocarbonos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL
12.
Chemosphere ; 155: 436-443, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27139122

RESUMO

Perfluorooctanoic acid (PFOA) and perfluorooctane sulfonate (PFOS) are two of the most widely used perfluoroalkyl acids (PFAAs). Because of their strong persistence, they have become widely distributed throughout the environment and human bodies. PFOA and PFOS are suspected to disrupt the endocrine system based upon many in vivo studies, but the underlying mechanisms are currently unclear. In this study, we investigated the endocrine-related effects of PFOA and PFOS using in vitro estrogen receptor (ER) and androgen receptor (AR) transactivation assays and steroidogenesis assay. The results showed that PFOA and PFOS exhibited weak antagonistic ER transactivation but did not exhibit agonistic ER or AR transactivation. In the steroidogenesis assay, PFOA and PFOS induced 17ß-estradiol (E2) level and reduced testosterone level, which would be caused by the induction of aromatase activity. The qPCR analysis of genes involved in steroidogenesis indicates that PFOA and PFOS associate with sex hormone synthesis by the transcriptional induction of two genes, cyp19 and 3ß-hsd2. Moreover, the transcriptional induction of cyp11b2 by PFOS suggests that this chemical may underlie the disruption of several physiological functions related to aldosterone. The results of the current study suggest that PFOA and PFOS are potential endocrine disrupting chemicals (EDCs) and provide information for further studies on the molecular events that initiate the adverse endocrine effects.


Assuntos
Carcinoma Adrenocortical/genética , Ácidos Alcanossulfônicos/farmacologia , Caprilatos/farmacologia , Disruptores Endócrinos/farmacologia , Sistema Endócrino/efeitos dos fármacos , Fluorocarbonos/farmacologia , Hormônios Esteroides Gonadais/metabolismo , Receptores Androgênicos/genética , Receptores de Estrogênio/genética , Carcinoma Adrenocortical/metabolismo , Carcinoma Adrenocortical/patologia , Aromatase/genética , Aromatase/metabolismo , Bioensaio , Estrogênios/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Reação em Cadeia da Polimerase em Tempo Real , Receptores Androgênicos/metabolismo , Receptores de Estrogênio/metabolismo , Células Tumorais Cultivadas
13.
Chemosphere ; 155: 488-497, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27151425

RESUMO

This study was undertaken to evaluate the possible role of several reproductive hormone receptors on the disruption of the hypothalamic-pituitary-testis (HPT) axis activity induced by perfluorooctane sulfonate (PFOS). The studied receptors are the gonadotropin-releasing hormone receptor (GnRHr), luteinizing hormone receptor (LHr), follicle-stimulating hormone receptor (FSHr), and the androgen receptor (Ar). Adult male rats were orally treated with 1.0; 3.0 and 6.0 mg of PFOS kg(-1) d(-1) for 28 days. In general terms, PFOS can modify the relative gene and protein expressions of these receptors in several tissues of the reproductive axis. At the testicular level, apart from the expected inhibition of both gene and protein expressions of FSHr and Ar, PFOS also stimulates the GnRHr protein and the LHr gene expression. The receptors of the main hormones involved in the HPT axis may have an important role in the disruption exerted by PFOS on this axis.


Assuntos
Ácidos Alcanossulfônicos/farmacologia , Fluorocarbonos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Receptores Androgênicos/metabolismo , Receptores do FSH/metabolismo , Receptores LHRH/metabolismo , Receptores do LH/metabolismo , Ácidos Alcanossulfônicos/química , Animais , Western Blotting , Fluorocarbonos/química , Hormônio Foliculoestimulante/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Hormônio Luteinizante/metabolismo , Masculino , Reação em Cadeia da Polimerase , Ratos , Ratos Sprague-Dawley , Receptores Androgênicos/genética , Receptores do FSH/genética , Receptores do LH/genética , Receptores LHRH/genética , Reprodução/efeitos dos fármacos , Testículo/efeitos dos fármacos , Testículo/metabolismo
14.
Toxicol Appl Pharmacol ; 295: 85-93, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26844784

RESUMO

Studies demonstrated that perfluorooctane sulfonate (PFOS) tends to accumulate in the liver and is capable to cause hepatomegaly. In the present study, we investigated the roles of miR-155 in PFOS-induced hepatotoxicity in SD rats and HepG2 cells. Male SD rats were orally administrated with PFOS at 1 or 10mg/kg/day for 28 days while HepG2 cells were treated with 0-50 µM of PFOS for 24h or 50 µM of PFOS for 1, 3, 6, 12 or 24h, respectively. We found that PFOS significantly increased the liver weight and serum alanine transaminase (ALT) and aspartate amino transferase (AST) levels in rats. Morphologically, PFOS caused actin filament remodeling and endothelial permeability changes in the liver. Moreover, PFOS triggered reactive oxygen species (ROS) generation and induced apoptosis in both in vivo and in vitro assays. Immunoblotting data showed that NF-E2-related factor-2 (Nrf2) expression and activation and its target genes were all suppressed by PFOS in the liver and HepG2 cells. However, PFOS significantly increased miR-155 expression. Further studies showed that pretreatment of HepG2 cells with catalase significantly decreased miR-155 expression and substantially increased Nrf2 expression and activation, resulting in reduction of PFOS-induced cytotoxicity and oxidative stress. Taken together, these results indicated that miR-155 plays an important role in the PFOS-induced hepatotoxicity by disrupting Nrf2/ARE signaling pathway.


Assuntos
Ácidos Alcanossulfônicos/farmacologia , Doença Hepática Induzida por Substâncias e Drogas/fisiopatologia , Fluorocarbonos/farmacologia , MicroRNAs/biossíntese , Fator 2 Relacionado a NF-E2/biossíntese , Citoesqueleto de Actina/metabolismo , Alanina Transaminase/metabolismo , Animais , Apoptose/efeitos dos fármacos , Aspartato Aminotransferases/metabolismo , Relação Dose-Resposta a Droga , Células Endoteliais/metabolismo , Células Hep G2 , Humanos , Masculino , Tamanho do Órgão , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo
15.
J Hazard Mater ; 305: 219-228, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26686481

RESUMO

Aquatic oligochaete Limnodrilus hoffmeisteri (L. hoffmeisteri) has been commonly used as a lethal and/or sub-lethal toxicological model organism in ecological risk assessments in contaminated water environments. In this study, experiments were conducted to investigate the potential toxic effects of copper (Cu(II)) with or without perfluorooctane sulfonate (PFOS) under different pH values (6.0, 7.0 and 8.0) on LC50, bioaccumulation, and oxidative stress biomarkers in L. hoffmeisteri after 3 and 7 days. The LC50 values of Cu(II) decreased with the increasing pH and the addition of PFOS. After each exposure, increasing bioaccumulation of Cu(II) in L. hoffmeisteri was observed in the combined exposure treatments, whereas the bioaccumulation of PFOS decreased. Moreover, the activity of superoxide dismutase, the level of glutathione, and the content of malondialdehyde were significantly altered after these exposures, possibly indicating that the bioaccumulation of Cu(II) and PFOS caused adverse effects on antioxidant defenses of L. hoffmeisteri. The integrated biomarker response index, indicates that the combined effect was proposed as synergism, which is coincided with the results of toxic unit. Moreover, this work showed that aquatic environment may become more livable when water conditions changed from acidic to near-neutral or alkaline.


Assuntos
Ácidos Alcanossulfônicos/toxicidade , Cobre/toxicidade , Fluorocarbonos/toxicidade , Oligoquetos/efeitos dos fármacos , Poluentes Químicos da Água/toxicidade , Ácidos Alcanossulfônicos/farmacologia , Animais , Cobre/farmacocinética , Fluorocarbonos/farmacologia , Glutationa/metabolismo , Concentração de Íons de Hidrogênio , Dose Letal Mediana , Malondialdeído/metabolismo , Oligoquetos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/metabolismo , Poluentes Químicos da Água/farmacocinética
16.
PLoS One ; 10(8): e0136829, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26313651

RESUMO

AIM: Triple-negative breast cancer (TNBC) is considered one of the most aggressive subtypes of breast cancer. Near infrared photoimmunotherapy (NIR-PIT) is a cancer treatment that employs an antibody-photosensitizer conjugate (APC) followed by exposure of NIR light for activating selective cytotoxicity on targeted cancer cells and may have application to TNBC. In order to minimize the dose of APC while maximizing the therapeutic effects, dosing of the APC and NIR light need to be optimized. In this study, we investigate in vitro and in vivo efficacy of cetuximab (cet)-IR700 NIR-PIT on two breast cancer models MDAMB231 (TNBC, EGFR moderate) and MDAMB468 (TNBC, EGFR high) cell lines, and demonstrate a method to optimize the dosing APC and NIR light. METHOD: After validating in vitro cell-specific cytotoxicity, NIR-PIT therapeutic effects were investigated in mouse models using cell lines derived from TNBC tumors. Tumor-bearing mice were separated into 4 groups for the following treatments: (1) no treatment (control); (2) 300 µg of cet-IR700 i.v., (APC i.v. only); (3) NIR light exposure only, NIR light was administered at 50 J/cm2 on day 1 and 100 J/cm2 on day 2 (NIR light only); (4) 300 µg of cet-IR700 i.v., NIR light was administered at 50 J/cm2 on day 1 after injection and 100 J/cm2 of light on day 2 after injection (one shot NIR-PIT). To compare different treatment regimens with a fixed dose of APC, we added the following treatments (5) 100 µg of cet-IR700 i.v., NIR light administered at 50 J/cm2 on day 1 and 50 µg of cet-IR700 i.v. immediately after NIR-PIT, then NIR light was administered at 100 J/cm2 on day 2, which were performed two times every week ("two split" NIR-PIT) and (6) 100 µg of cet-IR700 i.v., NIR light was administered at 50 J/cm2 on day 1 and 100 J/cm2 on day 2, which were performed three times per week ("three split" NIR-PIT). RESULT: Both specific binding and NIR-PIT effects were greater with MDAMB468 than MDAMB231 cells in vitro. Tumor accumulation of cet-IR700 in MDAMB468 tumors was significantly higher (p < 0.05) than in MDAMB231 tumors in vivo. Tumor growth and survival of MDAMB231 tumor bearing mice was significantly lower in the NIR-PIT treatment group (p < 0.05). In MDAMB468 bearing mice, tumor growth and survival was significantly improved in the NIR-PIT treatment groups in all treatment regimens (one shot NIR-PIT; p < 0.05, "two split" NIR-PIT; p < 0.01, "three split" NIR-PIT; p < 0.001) compared with control groups. CONCLUSION: NIR-PIT for TNBC was effective regardless of expression of EGFR, however, greater cell killing was shown with higher EGFR expression tumor in vitro. In all treatment regimens, NIR-PIT suppressed tumor growth, resulting in significantly prolonged survival that further improved by splitting the APC dose and using repeated light exposures.


Assuntos
Receptores ErbB/metabolismo , Imunoterapia/métodos , Fototerapia/métodos , Neoplasias de Mama Triplo Negativas/terapia , Ácidos Alcanossulfônicos/química , Ácidos Alcanossulfônicos/farmacologia , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Cetuximab/farmacologia , Feminino , Humanos , Indóis/química , Indóis/farmacologia , Luz , Camundongos Nus , Fármacos Fotossensibilizantes/farmacologia , Neoplasias de Mama Triplo Negativas/metabolismo , Neoplasias de Mama Triplo Negativas/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
17.
J Hazard Mater ; 299: 361-70, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26143199

RESUMO

Perfluorooctane sulfonate (PFOS) is a commonly used and widely distributed perfluorinated compound proven to cause adverse health outcomes. However, how PFOS affects liver cell proliferation is not well understood. In this experiment, we exposed a human liver cell line (HL-7702) to 50 µM PFOS for 48 h and 96 h. We identified 52 differentially expressed proteins using a quantitative proteomic approach. Among them, 27 were associated with cell proliferation, including hepatoma-derived growth factor (Hdgf) and proliferation biomarkers Mk167 (Ki67) and Top2α. Results from MTT, cell counting, and cell cycle analysis showed low-dose PFOS (<200 µM) stimulated HL-7702 cell viability at 48 h and 96 h, reduced the G0/G1 percentage, and increased the S+G2/M percentage. Moreover, levels of Cyclin D1, Cyclin E2, Cyclin A2, Cyclin B1 and their partner Cdks were elevated, and the expression of regulating proteins like c-Myc, p53, p21 waf/cip1 and Myt1, as well as the phosphorylation levels of p-Wee1(S642), p-Chk1(S345) and p-Chk2(T68), were disturbed. We hypothesized that low-dose PFOS stimulated HL-7702 proliferation by driving cells into G1 through elevating cyclins/cdks expression, and by promoting cell cycle progression through altering other regulating proteins. This research will shed light on the mechanisms behind PFOS-mediated human hepatotoxicity.


Assuntos
Ácidos Alcanossulfônicos/farmacologia , Proliferação de Células , Fluorocarbonos/farmacologia , Fígado/efeitos dos fármacos , Proteômica , Ciclo Celular , Linhagem Celular , Humanos , Fígado/citologia
18.
Chemosphere ; 129: 239-45, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25455676

RESUMO

Perfluoroalkylated substances (PFAS) have been widely used since 1950s and humans are exposed through food, drinking water, consumer products, dust, etc. The long-chained PFAS are persistent in the environment and accumulate in wildlife and humans. They are suspected carcinogens and a potential mode of action is through generation of oxidative stress. Seven long-chained PFAS found in human serum were investigated for the potential to generate reactive oxygen species (ROS), induce DNA damage and disturb the total antioxidant capacity (TAC). The tested PFAS were perfluorohexane sulfonate (PFHxS), perfluorooctane sulfonic acid (PFOS), perfluoroctanoic acid (PFOA), perfluorononanoate (PFNA), perfluorodecanoate (PFDA), perfluoroundecanoate (PFUnA), and perfluorododecanoate (PFDoA). Using the human hepatoma cell line (HepG2) and an exposure time of 24h we found that all three endpoints were affected by one or more of the compounds. PFHxS, PFOA, PFOS and PFNA showed a dose dependent increase in DNA damage in the concentration range from 2×10(-7) to 2×10(-5)M determined by the comet assay. Except for PFDoA, all the other PFAS increased ROS generation significantly. For PFHxS and PFUnA the observed ROS increases were dose-dependent. Cells exposed to PFOA were found to have a significant lower TAC compared with the solvent control, whereas a non-significant trend in TAC decrease was observed for PFOS and PFDoA and an increase tendency for PFHxS, PFNA and PFUnA. Our results indicate a possible genotoxic and cytotoxic potential of the PFAS in human liver cells.


Assuntos
Ácidos Alcanossulfônicos/farmacologia , Fluorocarbonos/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Alcanossulfonatos , Antioxidantes , Biomarcadores , Ensaio Cometa , Dano ao DNA , Humanos , Espécies Reativas de Oxigênio
19.
Photochem Photobiol ; 90(4): 845-52, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24597763

RESUMO

We previously demonstrated that the nonionic surfactants, nonylphenol polyethoxylates (NPEOs) induced the phosphorylation of histone H2AX (γ-H2AX), accompanied by DNA double-strand breaks (DSBs), and that exposure to ultraviolet (UV) degraded NPEOs, which sometimes enhanced their DNA-damaging ability. In this study, we showed that linear alkylbenzene sulfonates (LAS), general anion surfactants, also generated DSBs with γ-H2AX, and this ability was attenuated by UVB exposure. In the human breast adenocarcinoma cell line, MCF-7, γ-H2AX was generated in a dose-dependent manner immediately after cells were treated with LAS, and this was attributed to the formation of DSBs and was independent of cell cycle phases. The ability to generate γ-H2AX was markedly reduced in LAS exposed to UVB. HPLC analysis revealed that LAS were a mixture of various alkyl chain lengths, the peaks of which were detected at individual retention times. UVB evenly decreased all peaks of LAS, without migration of peaks to other retention times, which indicated that UVB may degrade the benzene ring of LAS, but did not shorten the alkyl chains. UVB is an important environmental factor in the degradation of LAS exhibiting the ability to induce DSBs, the most serious type of DNA damage.


Assuntos
Ácidos Alcanossulfônicos/farmacologia , Regulação da Expressão Gênica/efeitos da radiação , Histonas/metabolismo , Raios Ultravioleta , Adenocarcinoma/metabolismo , Histonas/química , Histonas/genética , Humanos , Células MCF-7 , Fosforilação
20.
Arch Pharm Res ; 37(8): 983-91, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24190754

RESUMO

In this study, we isolated a new sulfonic acid derivative, (Z)-4-methylundeca-1,9-diene-6-sulfonic acid (1), from the sea urchin collected from the Sea of Okhotsk. We established the structure of this new compound by analysis of NMR and HRMS data, along with comparison of the data with those of the related compounds reported in the literature. In addition, we investigated its anti-inflammatory effects in LPS-stimulated RAW264.7 macrophages. Compound 1 inhibited the production of NO, iNOS, PGE2, and COX-2, and it also suppressed the production of pro-inflammatory cytokines, such as TNF-α and IL-1ß. It inhibited the translocation of the NF-κB subunit p65 into the nucleus by interrupting the phosphorylation and degradation of IκB-α. In addition, compound 1 significantly decreased the phosphorylation of JNK and p38 in LPS-stimulated RAW264.7 macrophages, suggesting that suppression of the inflammation process by compound 1 was mediated through the MAPK pathway. Taken together, this study showed that the anti-inflammatory effects of a new sulfonic acid derivative, (Z)-4-methylundeca-1,9-diene-6-sulfonic acid were mediated through the inhibition of NF-κB and JNK/p38 MAPK signaling pathways.


Assuntos
Ácidos Alcanossulfônicos/farmacologia , Anti-Inflamatórios/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , NF-kappa B/antagonistas & inibidores , Ouriços-do-Mar/química , Ácidos Sulfônicos/farmacologia , Ácidos Alcanossulfônicos/efeitos adversos , Ácidos Alcanossulfônicos/isolamento & purificação , Animais , Anti-Inflamatórios/efeitos adversos , Anti-Inflamatórios/isolamento & purificação , Western Blotting , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Temperatura Baixa , MAP Quinase Quinase 4/antagonistas & inibidores , Macrófagos/enzimologia , Macrófagos/imunologia , Camundongos , Óxido Nítrico/metabolismo , Oceanos e Mares , Ouriços-do-Mar/crescimento & desenvolvimento , Ácidos Sulfônicos/efeitos adversos , Ácidos Sulfônicos/isolamento & purificação , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA