Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Chemosphere ; 350: 140975, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38142884

RESUMEN

Nanomaterials (NMs) are defined as materials with at least one external dimension below 100 nm. Their small size confers them interesting unique physico-chemical properties, hence NMs are increasingly used in a diversity of applications. However, the specific properties of NMs could also make them more harmful than their bulk counterparts. Therefore, there is a crucial need to deliver efficient NM hazard assessment in order to sustain the responsible development of nanotechnology. This study analysed the genotoxic potential of several NMs: one titanium dioxide (TiO2) and two zinc oxide NMs (ZnO) that were tested up to 100 µg/mL on 2D and 3D hepatic HepaRG models. Genotoxicity analysis was performed comparing the alkaline comet assay in classical and high throughput formats. Moreover, oxidative DNA lesions were investigated with the Fpg-modified comet assay. Results showed that TiO2 NMs were not cytotoxic and not genotoxic in either cell model, although a small increase in the % tail DNA was observed in 3D HepaRG cells at 100 µg/mL in the classical format. The two ZnO NMs (ZnO S. NMs a commercial suspension and NM110 provided by the European Union Joint Research Centre) induced a concentration-dependent increase in cytotoxicity that was more pronounced in the 2D (>20% cytotoxicity was observed for ZnO S. at concentrations greater than 25 µg/mL, and for NM 110 at 50 µg/mL) than in the 3D model (more than 20% cytotoxicity for ZnO S. NMs at 50 µg/mL). While ZnO S. NMs induced DNA damage associated with cytotoxicity (at 25 and 50 µg/mL in 2D and 50 µg/mL in 3D), NM110 showed a clear genotoxic effect at non-cytotoxic concentrations (25 µg/mL in 2D and at 25 and 50 µg/mL in 3D). No major differences could be observed in the comet assay in the presence or absence of the Fpg enzyme. High throughput analysis using CometChip® mostly confirmed the results obtained with the classical format, and even enhanced the detection of genotoxicity in the 3D model. In conclusion, this study demonstrated that new approach methodologies (NAMs), 3D models and the high throughput format for the comet assay, were more efficient in the detection of genotoxic effects, and are therefore promising approaches to improve hazard assessment of NMs.


Asunto(s)
Óxido de Zinc , Ensayo Cometa/métodos , Óxido de Zinc/toxicidad , Daño del ADN , Oxidación-Reducción , Hígado
2.
Front Toxicol ; 5: 1220998, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37492623

RESUMEN

Carcinogenic chemicals, or their metabolites, can be classified as genotoxic or non-genotoxic carcinogens (NGTxCs). Genotoxic compounds induce DNA damage, which can be detected by an established in vitro and in vivo battery of genotoxicity assays. For NGTxCs, DNA is not the primary target, and the possible modes of action (MoA) of NGTxCs are much more diverse than those of genotoxic compounds, and there is no specific in vitro assay for detecting NGTxCs. Therefore, the evaluation of the carcinogenic potential is still dependent on long-term studies in rodents. This 2-year bioassay, mainly applied for testing agrochemicals and pharmaceuticals, is time-consuming, costly and requires very high numbers of animals. More importantly, its relevance for human risk assessment is questionable due to the limited predictivity for human cancer risk, especially with regard to NGTxCs. Thus, there is an urgent need for a transition to new approach methodologies (NAMs), integrating human-relevant in vitro assays and in silico tools that better exploit the current knowledge of the multiple processes involved in carcinogenesis into a modern safety assessment toolbox. Here, we describe an integrative project that aims to use a variety of novel approaches to detect the carcinogenic potential of NGTxCs based on different mechanisms and pathways involved in carcinogenesis. The aim of this project is to contribute suitable assays for the safety assessment toolbox for an efficient and improved, internationally recognized hazard assessment of NGTxCs, and ultimately to contribute to reliable mechanism-based next-generation risk assessment for chemical carcinogens.

3.
Nutrients ; 15(9)2023 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-37432249

RESUMEN

Docosahexaenoic acid (DHA, C22:6 ω-3) is a dietary polyunsaturated fatty acid that has an important role in human health. Epidemiological studies linked a high intake of DHA to a reduced risk of certain cancers. Recently, attention focused on how the lipid carrier in which DHA is delivered, i.e., esterified on acylglycerols, phospholipids, or free, affects its biological effects. However, studies comparing the effects of these different forms for DHA supply to cancer cells in vitro are limited. In this study, the effect of free DHA and five lipids carrying one to three DHA chains (LPC-DHA, PC-DHA, MAG-DHA, DAG-DHA and TAG-DHA) on the viability of the MDA-MB-231 breast cancer cell line was compared. Our results revealed a strong structure-function relationship of DHA-carrying lipids on the viability of MDA-MB-231 cells. Glycerophosphocholine-based lipids are the most effective DHA carriers in reducing the viability of MDA-MB-231 cells, with LPC-DHA being more effective (IC50 = 23.7 µM) than PC-DHA (IC50 = 67 µM). The other tested lipids are less toxic (MAG-DHA, free DHA) or even not toxic (DAG-DHA, TAG-DHA) under our conditions. Investigating the mechanism of cell death induced by LPC-DHA revealed increased oxidative stress and membrane cell damage.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama , Humanos , Femenino , Lisofosfatidilcolinas/farmacología , Ácidos Docosahexaenoicos/farmacología , Ácidos Grasos , Células MDA-MB-231
4.
Int J Mol Sci ; 24(9)2023 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-37175512

RESUMEN

This study investigates the genotoxicity and cytotoxicity of C17-sphinganine analog mycotoxin (C17-SAMT) using in vitro assays. C17-SAMT was previously identified as the cause of unusual toxicity in cultured mussels from the Bizerte Lagoon in northern Tunisia. While a previous in vivo genotoxicity study was inconclusive, in vitro results demonstrated that C17-SAMT induced an increase in micronucleus formation in human lymphoblastoid TK6 cells at concentrations of 0.87 µM and 1.74 µM. In addition, multiparametric cytotoxicity assays were performed in the human hepatoma HepaRG cell line, which showed that C17-SAMT induced mitochondrial dysfunction, decreased cellular ATP levels, and altered the expression of various proteins, including superoxide dismutase SOD2, heme oxygenase HO-1, and NF-κB. These results suggest that C17-SAMT is mutagenic in vitro and can induce mitochondrial dysfunction in HepaRG cells. However, the exact mode of action of this toxin requires further investigation. Overall, this study highlights the potential toxicity of C17-SAMT and the need for further research to better understand its effects.


Asunto(s)
Micotoxinas , Humanos , Línea Celular , Mutágenos/toxicidad , Toxinas Marinas/toxicidad , Daño del ADN , Pruebas de Micronúcleos/métodos
5.
Part Fibre Toxicol ; 19(1): 37, 2022 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-35578293

RESUMEN

BACKGROUND: TiO2 nanomaterials (NMs) are present in a variety of food and personal hygiene products, and consumers are exposed daily to these NMs through oral exposition. While the bulk of ingested TiO2 NMs are eliminated rapidly in stool, a fraction is able to cross the intestinal epithelial barrier and enter systemic circulation from where NMs can be distributed to tissues, primarily liver and spleen. Daily exposure to TiO2 NMs, in combination with a slow rate of elimination from tissues, results in their accumulation within different tissues. Considerable evidence suggests that following oral exposure to TiO2 NMs, the presence of NMs in tissues is associated with a number of adverse effects, both in intestine and liver. Although numerous studies have been performed in vitro investigating the acute effects of TiO2 NMs in intestinal and hepatic cell models, considerably less is known about the effect of repeated exposure on these models. In this study, we investigated the cytotoxic effects of repeated exposure of relevant models of intestine and liver to two TiO2 NMs differing in hydrophobicity for 24 h, 1 week and 2 weeks at concentrations ranging from 0.3 to 80 µg/cm2. To study the persistence of these two NMs in cells, we included a 1-week recovery period following 24 h and 1-week treatments. Cellular uptake by TEM and ToF-SIMS analyses, as well as the viability and pro-inflammatory response were evaluated. Changes in the membrane composition in Caco-2 and HepaRG cells treated with TiO2 NMs for up to 2 weeks were also studied. RESULTS: Despite the uptake of NM-103 and NM-104 in cells, no significant cytotoxic effects were observed in either Caco-2 or HepaRG cells treated for up to 2 weeks at NM concentrations up to 80 µg/cm2. In addition, no significant effects on IL-8 secretion were observed. However, significant changes in membrane composition were observed in both cell lines. Interestingly, while most of these phospholipid modifications were reversed following a 1-week recovery, others were not affected by the recovery period. CONCLUSION: These findings indicate that although no clear effects on cytotoxicity were observed following repeated exposure of differentiated Caco-2 and HepaRG cells to TiO2 NMs, subtle effects on membrane composition could induce potential adverse effects in the long-term.


Asunto(s)
Nanoestructuras , Titanio , Células CACO-2 , Hepatocitos , Humanos , Intestinos , Hígado , Nanoestructuras/toxicidad , Titanio/toxicidad
6.
Toxicol In Vitro ; 78: 105257, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34688838

RESUMEN

Exposure of consumers to aluminum-containing nanomaterials (Al NMs) is an area of concern for public health agencies. As the available data on the genotoxicity of Al2O3 and Al0 NMs are inconclusive or rare, the present study investigated their in vitro genotoxic potential in intestinal and liver cell models, and compared with the ionic form AlCl3. Intestinal Caco-2 and hepatic HepaRG cells were exposed to Al0 and Al2O3 NMs (0.03 to 80 µg/cm2). Cytotoxicity, oxidative stress and apoptosis were measured using High Content Analysis. Genotoxicity was investigated through γH2AX labelling, the alkaline comet and micronucleus assays. Moreover, oxidative DNA damage and carcinogenic properties were assessed using the Fpg-modified comet assay and the cell transforming assay in Bhas 42 cells respectively. The three forms of Al did not induce chromosomal damage. However, although no production of oxidative stress was detected, Al2O3 NMs induced oxidative DNA damage in Caco-2 cells but not likely related to ion release in the cell media. Considerable DNA damage was observed with Al0 NMs in both cell lines in the comet assay, likely due to interference with these NMs. No genotoxic effects were observed with AlCl3. None of the Al compounds induced cytotoxicity, apoptosis, γH2AX or cell transformation.


Asunto(s)
Aluminio/toxicidad , Daño del ADN , Nanopartículas del Metal/toxicidad , Cloruro de Aluminio/toxicidad , Óxido de Aluminio/toxicidad , Células CACO-2 , Línea Celular , Ensayo Cometa , Hepatocitos/efectos de los fármacos , Humanos , Intestinos/efectos de los fármacos , Pruebas de Micronúcleos , Estrés Oxidativo
7.
Toxicol In Vitro ; 73: 105125, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33631200

RESUMEN

Portimine, a recently identified cyclic imine produced by the dinoflagellate Vulcanodinium rugosum, has been described as a potent apoptotic agent in contrast to most of the cyclic imines that are well-known to be neurological toxins. As apoptosis can be a consequence of a high level of DNA lesions, we investigated the responses of portimine on several endpoints aimed at detecting DNA damage in the hepatic cell line HepaRG. Portimine induced phosphorylation of H2AX, which could possibly be consistent with the previously published induction of apoptosis with this toxin. In addition, detection of apoptosis through the activation of caspase-3, the induction of strand breaks detected by the comet assay as well as chromosome and genome mutations using the micronucleus assay were addressed. Surprisingly, portimine treatment resulted in increases in only γH2AX in differentiated HepaRG cells whereas no effects on the other endpoints were detected. These increases in γH2AX in the absence of genotoxic effects in the other tests could indicate that portimine could possibly induce a DNA replication stress and/or that the compound can be detoxified by the HepaRG cells.


Asunto(s)
Iminas/toxicidad , Toxinas Marinas/toxicidad , Compuestos de Espiro/toxicidad , Apoptosis/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Ensayo Cometa , Daño del ADN , Dinoflagelados , Histonas/metabolismo , Humanos , Hígado/citología , Pruebas de Micronúcleos
8.
Nanomaterials (Basel) ; 10(2)2020 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-32053952

RESUMEN

Due to several gaps remaining in the toxicological evaluation of nanomaterials (NMs), consumers and public health agencies have shown increasing concern for human health protection. In addition to aluminum (Al) microparticles, Al-containing nanomaterials (Al NMs) have been applied by food industry as additives and contact materials. Due to the limited amount of literature on the toxicity of Al NMs, this study aimed to evaluate the in vivo genotoxic potential of Al0 and Al2O3 NMs after acute oral exposure. Male Sprague-Dawley rats were administered three successive gavages at 6, 12.5 and 25 mg/kg bw. A comparison with AlCl3 was done in order to assess the potential effect of dissolution into Al ions. Both DNA strand breaks and oxidative DNA damage were investigated in six organs/tissues (duodenum, liver, kidney, spleen, blood and bone marrow) with the alkaline and the Fpg-modified comet assays. Concomitantly, chromosomal damage was investigated in bone marrow and colon with the micronucleus assay. The comet assay only showed DNA damage with Al2O3 NMs in bone marrow (BM), while AlCl3 induced slight but non-significant oxidative DNA damage in blood. No increase of chromosomal mutations was observed after treatment with the two Al MNs either in the BM or in the colons of rats.

9.
Nanotoxicology ; 13(7): 909-922, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30938204

RESUMEN

Aluminum (Al) can be ingested from food and released from packaging and can reach key organs involved in human metabolism, including the liver via systemic distribution. Recent studies discuss the occurrence of chemically distinct Al-species and their interconversion by contact with biological fluids. These Al species can vary with regard to their intestinal uptake, systemic transport, and therefore could have species-specific effects on different organs and tissues. This work aims to assess the in vitro hepatotoxic hazard potential of three different relevant Al species: soluble AlCl3 and two nanoparticulate Al species were applied, representing for the first time an investigation of metallic nanoparticles besides to mineral bound γ-Al2O3 on hepatic cell lines. To investigate the uptake and toxicological properties of the Al species, we used two different human hepatic cell lines: HepG2 and differentiated HepaRG cells. Cellular uptake was determined by different methods including light microscopy, transmission electron microscopy, side-scatter analysis, and elemental analysis. Oxidative stress, mitochondrial dysfunction, cell death mechanisms, and DNA damage were monitored as cellular parameters. While cellular uptake into hepatic cell lines occurred predominantly in the particle form, only ionic AlCl3 caused cellular effects. Since it is known, that Al species can convert one into another, and mechanisms including 'trojan-horse'-like uptake can lead to an Al accumulation in the cells. This could result in the slow release of Al ions, for which reason further hazard cannot be excluded. Therefore, individual investigation of the different Al species is necessary to assess the toxicological potential of Al particles.


Asunto(s)
Cloruro de Aluminio/toxicidad , Óxido de Aluminio/toxicidad , Daño del ADN , Hígado/efectos de los fármacos , Nanopartículas del Metal/toxicidad , Estrés Oxidativo/efectos de los fármacos , Cloruro de Aluminio/metabolismo , Óxido de Aluminio/metabolismo , Transporte Biológico , Supervivencia Celular/efectos de los fármacos , Células Hep G2 , Humanos , Hígado/metabolismo , Microscopía Electrónica de Transmisión
10.
Nanotoxicology ; 12(9): 992-1013, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30317887

RESUMEN

Aluminum (Al) is one of the most common elements in the earth crust and increasingly used in food, consumer products and packaging. Its hazard potential for humans is still not completely understood. Besides the metallic form, Al also exists as mineral, including the insoluble oxide, and in soluble ionic forms. Representatives of these three species, namely a metallic and an oxidic species of Al-containing nanoparticles and soluble aluminum chloride, were applied to human intestinal cell lines as models for the intestinal barrier. We characterized physicochemical particle parameters, protein corona composition, ion release and cellular uptake. Different in vitro assays were performed to determine potential effects and molecular modes of action related to the individual chemical species. For a deeper insight into signaling processes, microarray transcriptome analyses followed by bioinformatic data analysis were employed. The particulate Al species showed different solubility in biological media. Metallic Al nanoparticles released more ions than Al2O3 nanoparticles, while AlCl3 showed a mixture of dissolved and agglomerated particulate entities in biological media. The protein corona composition differed between both nanoparticle species. Cellular uptake, investigated in transwell experiments, occurred predominantly in particulate form, whereas ionic Al was not taken up by intestinal cell lines. Transcellular transport was not observed. None of the Al species showed cytotoxic effects up to 200 µg Al/mL. The transcriptome analysis indicated mainly effects on oxidative stress pathways, xenobiotic metabolism and metal homeostasis. We have shown for the first time that intestinal cellular uptake of Al occurs preferably in the particle form, while toxicological effects appear to be ion-related.


Asunto(s)
Aluminio/toxicidad , Mucosa Intestinal/efectos de los fármacos , Nanopartículas del Metal/toxicidad , Corona de Proteínas/metabolismo , Transcriptoma/efectos de los fármacos , Aluminio/química , Aluminio/metabolismo , Apoptosis/efectos de los fármacos , Transporte Biológico , Células CACO-2 , Supervivencia Celular/efectos de los fármacos , Humanos , Mucosa Intestinal/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Nanopartículas del Metal/química , Propiedades de Superficie
11.
Artículo en Inglés | MEDLINE | ID: mdl-29076405

RESUMEN

Food contact paperboards may be a potential source of food contamination as they can release chemicals (intentionally added or not), especially recycled paperboards. This study assessed the in vitro genotoxicity of food contact paperboard samples from a manufacturer, collected at the beginning and at the end of a recycling production chain. Samples were extracted in water to mimic a wet food contact. Different genotoxic endpoints were evaluated in two human hepatic cell lines (HepG2 and HepaRG) using bioassays: γH2AX and p53 activation, primary DNA damage with the comet assay and micronucleus formation. It was found that the samples from the beginning and the end of the production chain induced, with the same potency, γH2AX and p53-ser15 activation and DNA damage with the comet assay. The micronucleus assay was negative with the paperboard extract from the beginning of the chain, whereas positive data were observed for the end paperboard extract. These results indicate that samples from recycled food contact paperboard can induce in vitro genotoxic effects in this study's experimental conditions.


Asunto(s)
Contaminación de Alimentos/análisis , Embalaje de Alimentos , Papel , Daño del ADN/efectos de los fármacos , Células Hep G2 , Histonas/genética , Humanos , Reciclaje , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética
12.
Artículo en Inglés | MEDLINE | ID: mdl-27273980

RESUMEN

With the growing numbers of nanomaterials (NMs), there is a great demand for rapid and reliable ways of testing NM safety-preferably using in vitro approaches, to avoid the ethical dilemmas associated with animal research. Data are needed for developing intelligent testing strategies for risk assessment of NMs, based on grouping and read-across approaches. The adoption of high throughput screening (HTS) and high content analysis (HCA) for NM toxicity testing allows the testing of numerous materials at different concentrations and on different types of cells, reduces the effect of inter-experimental variation, and makes substantial savings in time and cost. HTS/HCA approaches facilitate the classification of key biological indicators of NM-cell interactions. Validation of in vitro HTS tests is required, taking account of relevance to in vivo results. HTS/HCA approaches are needed to assess dose- and time-dependent toxicity, allowing prediction of in vivo adverse effects. Several HTS/HCA methods are being validated and applied for NM testing in the FP7 project NANoREG, including Label-free cellular screening of NM uptake, HCA, High throughput flow cytometry, Impedance-based monitoring, Multiplex analysis of secreted products, and genotoxicity methods-namely High throughput comet assay, High throughput in vitro micronucleus assay, and γH2AX assay. There are several technical challenges with HTS/HCA for NM testing, as toxicity screening needs to be coupled with characterization of NMs in exposure medium prior to the test; possible interference of NMs with HTS/HCA techniques is another concern. Advantages and challenges of HTS/HCA approaches in NM safety are discussed. WIREs Nanomed Nanobiotechnol 2017, 9:e1413. doi: 10.1002/wnan.1413 For further resources related to this article, please visit the WIREs website.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento/métodos , Nanoestructuras/toxicidad , Pruebas de Toxicidad/métodos , Animales , Línea Celular , Técnicas Citológicas , Humanos , Espacio Intracelular/química , Espacio Intracelular/metabolismo , Ratones
13.
Toxicol In Vitro ; 29(2): 398-407, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25448807

RESUMEN

Silica (SiO2) in its nanosized form is now used in food applications although the potential risks for human health need to be evaluated in further detail. In the current study, the uptake of 15 and 55nm colloidal SiO2 NPs in the human intestinal Caco-2 cell line was investigated by transmission electron microscopy. The ability of these NPs to induce cytotoxicity (XTT viability test), genotoxicity (γH2Ax and micronucleus assay), apoptosis (caspase 3), oxidative stress (oxidation of 2,7-dichlorodihydrofluorescein diacetate probe) and proinflammatory effects (interleukin IL-8 secretion) was evaluated. Quartz DQ12 was used as particle control. XTT and cytokinesis-block micronucleus assays revealed size- and concentration-dependent effects on cell death and chromosome damage following exposure to SiO2 nanoparticles, concomitantly with generation of reactive oxygen species (ROS), SiO2-15nm particles being the most potent. In the same way, an increased IL-8 secretion was only observed with SiO2-15nm at the highest tested dose (32µg/ml). TEM images showed that both NPs were localized within the cytoplasm but did not enter the nucleus. SiO2-15nm, and to a lower extent SiO2-55nm, exerted toxic effects in Caco-2 cells. The observed genotoxic effects of these NPs are likely to be mediated through oxidative stress rather than a direct interaction with the DNA. Altogether, our results indicate that exposure to SiO2 NPs may induce potential adverse effects on the intestinal epithelium in vivo.


Asunto(s)
Mutágenos/toxicidad , Nanopartículas/toxicidad , Dióxido de Silicio/toxicidad , Apoptosis/efectos de los fármacos , Células CACO-2 , Caspasa 3/metabolismo , Supervivencia Celular/efectos de los fármacos , Citoplasma/metabolismo , Histonas/metabolismo , Humanos , Interleucina-8/metabolismo , Pruebas de Micronúcleos , Microscopía Electrónica de Transmisión , Nanopartículas/ultraestructura , Estrés Oxidativo , Tamaño de la Partícula
14.
Mar Drugs ; 12(8): 4616-34, 2014 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-25196936

RESUMEN

The phycotoxin, okadaic acid (OA) and dinophysistoxin 1 and 2 (DTX-1 and -2) are protein phosphatase PP2A and PP1 inhibitors involved in diarrhetic shellfish poisoning (DSP). Data on the toxicity of the OA-group toxins show some differences with respect to the in vivo acute toxicity between the toxin members. In order to investigate whether OA and congeners DTX-1 and -2 may induce different mechanisms of action during acute toxicity on the human intestine, we compared their toxicological effects in two in vitro intestinal cell models: the colorectal adenocarcinoma cell line, Caco-2, and the intestinal muco-secreting cell line, HT29-MTX. Using a high content analysis approach, we evaluated various cytotoxicity parameters, including apoptosis (caspase-3 activation), DNA damage (phosphorylation of histone H2AX), inflammation (translocation of NF-κB) and cell proliferation (Ki-67 production). Investigation of the kinetics of the cellular responses demonstrated that the three toxins induced a pro-inflammatory response followed by cell cycle disruption in both cell lines, leading to apoptosis. Our results demonstrate that the three toxins induce similar effects, as no major differences in the cytotoxic responses could be detected. However DTX-1 induced cytotoxic effects at five-fold lower concentrations than for OA and DTX-2.


Asunto(s)
Intestinos/efectos de los fármacos , Ácido Ocadaico/toxicidad , Toxinas Biológicas/toxicidad , Apoptosis/efectos de los fármacos , Células CACO-2 , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Células HT29 , Humanos , Inflamación/metabolismo , Mucosa Intestinal/metabolismo , Piranos/toxicidad
15.
Mar Drugs ; 11(9): 3350-71, 2013 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-24002102

RESUMEN

Pinnatoxin G (PnTX-G) is a marine toxin belonging to the class of cyclic imines and produced by the dinoflagellate Vulcanodinium rugosum. In spite of its strong toxicity to mice, leading to the classification of pinnatoxins into the class of "fast-acting toxins", its hazard for human health has never been demonstrated. In this study, crude extracts of V. rugosum exhibited significant cytotoxicity against Neuro2A and KB cells. IC50 values of 0.38 µg mL⁻¹ and 0.19 µg mL⁻¹ were estimated on Neuro2A cells after only 24 h of incubation and on KB cells after 72 h of incubation, respectively. In the case of Caco-2 cells 48 h after exposure, the crude extract of V. rugosum induced cell cycle arrest accompanied by a dramatic increase in double strand DNA breaks, although only 40% cytotoxicity was observed at the highest concentration tested (5 µg mL⁻¹). However, PnTX-G was not a potent cytotoxic compound as no reduction of the cell viability was observed on the different cell lines. Moreover, no effects on the cell cycle or DNA damage were observed following treatment of undifferentiated Caco-2 cells with PnTX-G. The crude extract of V. rugosum was thus partially purified using liquid-liquid partitioning and SPE clean-up. In vitro assays revealed strong activity of some fractions containing no PnTX-G. The crude extract and the most potent fraction were evaluated using full scan and tandem high resolution mass spectrometry. The dereplication revealed the presence of a major compound that could be putatively annotated as nakijiquinone A, N-carboxy-methyl-smenospongine or stachybotrin A, using the MarinLit™ database. Further investigations will be necessary to confirm the identity of the compounds responsible for the cytotoxicity and genotoxicity of the extracts of V. rugosum.


Asunto(s)
Alcaloides/química , Alcaloides/farmacología , Dinoflagelados/química , Toxinas Marinas/química , Toxinas Marinas/farmacología , Compuestos de Espiro/química , Compuestos de Espiro/farmacología , Células CACO-2 , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Roturas del ADN de Doble Cadena/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Humanos , Células KB
16.
J Biochem Mol Toxicol ; 27(5): 253-8, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23554253

RESUMEN

While MC-LR and MC-RR share significant structural similarity, MC-RR is less cytotoxic than MC-LR. In the current study, we have compared the effects of MC-LR and MC-RR in Caco-2 cells by evaluating cytotoxicity, oxidative stress (reactive oxygen species production), and the cellular proinflammatory response (IL-6 and IL-8 production). Following treatment with 100 µM microcystins (MC), cytotoxicity was two-fold greater with MC-LR as compared to MC-RR after 24 h exposure. Whereas the reactive oxygen species production and IL-6 secretion were similar following a 24-h treatment with either MC, 100 µM MC-LR induced a five-fold greater IL-8 secretion when compared to MC-RR. Our study has demonstrated that, although both MC-LR and MC-RR induced some cytotoxicity in human intestinal cells, a major difference in IL-8 production was observed between the two variants.


Asunto(s)
Supervivencia Celular/efectos de los fármacos , Citocinas/metabolismo , Microcistinas/farmacología , Estrés Oxidativo/efectos de los fármacos , Células CACO-2 , Diferenciación Celular/efectos de los fármacos , Colorantes , Enterocitos/efectos de los fármacos , Humanos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Toxinas Marinas , Rojo Neutro , Especies Reactivas de Oxígeno/metabolismo , Sales de Tetrazolio
17.
Toxicol Sci ; 133(2): 289-97, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23535362

RESUMEN

Epidemiological studies indicate a role of genetic and environmental factors in Parkinson's disease involving alterations of the neuronal α-synuclein (α-syn) protein. In particular, a relationship between Parkinson's disease and occupational exposure to pesticides has been repeatedly suggested. Our objective was to precisely assess changes in α-syn levels in human neuroblastoma (SH-SY5Y) and melanoma (SK-MEL-2) cell lines following acute exposure to pesticides (rotenone, paraquat, maneb, and glyphosate) using Western blot and flow cytometry. These human cell lines express α-syn endogenously, and overexpression of α-syn (wild type or mutated A53T) can be obtained following recombinant adenoviral transduction. We found that endogenous α-syn levels in the SH-SY5Y neuroblastoma cell line were markedly increased by paraquat, and to a lesser extent by rotenone and maneb, but not by glyphosate. Rotenone also clearly increased endogenous α-syn levels in the SK-MEL-2 melanoma cell line. In the SH-SY5Y cell line, similar differences were observed in the α-syn adenovirus-transduced cells, with a higher increase of the A53T mutated protein. Paraquat markedly increased α-syn in the SK-MEL-2 adenovirus-transduced cell line, similarly for the wild-type or A53T proteins. The observed differences in the propensities of pesticides to increase α-syn levels are in agreement with numerous reports that indicate a potential role of exposure to certain pesticides in the development of Parkinson's disease. Our data support the hypothesis that pesticides can trigger some molecular events involved in this disease and also in malignant melanoma that consistently shows a significant but still unexplained association with Parkinson's disease.


Asunto(s)
Insecticidas/toxicidad , Melanoma/tratamiento farmacológico , Neuroblastoma/tratamiento farmacológico , Rotenona/toxicidad , alfa-Sinucleína/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Glicina/análogos & derivados , Glicina/toxicidad , Humanos , Maneb/toxicidad , Melanoma/metabolismo , Neuroblastoma/metabolismo , Paraquat/toxicidad , Enfermedad de Parkinson/etiología , Transducción Genética , Glifosato
18.
J Neurosci Methods ; 192(2): 268-76, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20709102

RESUMEN

Analyses using antibodies directed against α-synuclein play a key role in the understanding of the pathologies associated with neurodegenerative disorders such as Parkinson's disease (PD), dementia with Lewy bodies (DLB) and multiple system atrophy (MSA). However, the generation of antibodies against immunogens with significant sequence similarity to host proteins such as α-synuclein is often hindered by host immunotolerance. In contrast to wild-type C57BL/6J and BALB/c mice immunized with recombinant human α-synuclein, C57BL/6S Δsnca mice presenting a natural deletion of the α-synuclein locus, bypassed the immunotolerance process which resulted in a much higher polyclonal antibody response. The native or fibrillized conformation of α-synuclein used as the immunogen did not have an impact on the amounts of specific antibodies in sera of the host. The immunization protocols resulted in the generation of the IgG AS11, raised against fibrillized recombinant human α-synuclein in C57BL/6S Δsnca mice. This monoclonal antibody, recognizing an N-terminal α-synuclein epitope, was selected for its specificity and significant reactivity in Western-blot, immunofluorescence and immunohistochemistry assays. The ability of AS11 to detect both soluble and aggregated forms of α-synuclein present in pathological cytoplasmic inclusions was further assessed using analysis of human brains with PD or MSA, transgenic mouse lines expressing A53T human α-synuclein, and cellular models expressing human α-synuclein. Taken together, our study indicates that novel antibodies helpful to characterize alterations of α-synuclein leading to neurodegeneration in PD and related disorders could be efficiently developed using this original immunization strategy.


Asunto(s)
Anticuerpos Monoclonales/biosíntesis , alfa-Sinucleína/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Western Blotting , Línea Celular , Células Cultivadas , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , alfa-Sinucleína/genética
19.
Mol Cell Endocrinol ; 316(1): 53-9, 2010 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-19786070

RESUMEN

Sex hormone-binding globulin (SHBG) is the main transport binding protein for sex steroid hormones in plasma and regulates their accessibility to target cells. Plasma SHBG is secreted by the liver under the control of hormones and nutritional factors. In the human hepatoma cell line (HepG2), thyroid and estrogenic hormones, and a variety of drugs including the antioestrogen tamoxifen, the phytoestrogen, genistein and mitotane (Op'DDD) increase SHBG production and SHBG gene promoter activity. In contrast, monosaccharides (glucose or fructose) effectively decrease SHBG expression by inducing lipogenesis, which reduces hepatic HNF-4alpha levels, a transcription factor that play a critical role in controlling the SHBG promoter. Interestingly, diminishing hepatic lipogenesis and free fatty acid liver biosynthesis also appear to be associated with the positive effects of thyroid hormones and PPARgamma antagonists on SHBG expression. This mechanism provides a biological explanation for why SHBG is a sensitive biomarker of insulin resistance and the metabolic syndrome, and why low plasma SHBG levels are a risk factor for developing hyperglycemia and type 2 diabetes, especially in women. These important advances in our knowledge of the regulation of SHBG expression in the liver open new approaches for identifying and preventing metabolic disorder-associated diseases early in life.


Asunto(s)
Hígado/metabolismo , Síndrome Metabólico/metabolismo , Globulina de Unión a Hormona Sexual/metabolismo , Empalme Alternativo , Animales , Diabetes Mellitus Tipo 2/metabolismo , Hormonas Esteroides Gonadales/metabolismo , Humanos , Insulina/metabolismo , Monosacáridos/metabolismo , PPAR gamma/metabolismo , Globulina de Unión a Hormona Sexual/genética , Hormonas Tiroideas/metabolismo
20.
Blood ; 113(7): 1444-54, 2009 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-18952892

RESUMEN

The human Mixed-Lineage-Leukemia-5 (MLL5) gene is located in a genomic region frequently deleted in patients with myeloid malignancies and encodes a widely expressed nuclear protein most closely related to MLL1, a Trithorax transcriptional regulator with established involvement in leukemogenesis. Although the physiologic function of MLL5 is completely unknown, domain structure and homology to transcriptional regulators with histone methyltransferase activity suggest a role in epigenetic gene regulation. To investigate physiologic functions of Mll5, we have generated a knockout mouse mutant using Cre/loxP technology. Adult homozygous Mll5-deficient mice are obtained at reduced frequency because of postnatal lethality. Surviving animals display a variety of abnormalities, including male infertility, retarded growth, and defects in multiple hematopoietic lineages. Interestingly, Mll5(-/-) mice die of sublethal whole-body irradiation but can be rescued with wild-type bone marrow grafts. Flow cytometric ana-lysis, bone marrow reconstitution, and in vivo BrdU-labeling experiments reveal numerical, functional, and cell-cycle defects in the lineage-negative Sca-1(+), Kit(+) (LSK) population, which contains short- and long-term hematopoietic stem cells. Together, these in vivo findings establish several nonredundant functions for Mll5, including an essential role in regulating proliferation and functional integrity of hematopoietic stem/progenitor cells.


Asunto(s)
Trastornos del Crecimiento/genética , Hematopoyesis/inmunología , Células Madre Hematopoyéticas/citología , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Animales , Diferenciación Celular/inmunología , Femenino , Genes Letales , Trastornos del Crecimiento/inmunología , Heterocigoto , Infertilidad Masculina/genética , Infertilidad Masculina/inmunología , Linfocitos/citología , Masculino , Ratones , Ratones Noqueados , Fenotipo , Embarazo , Tolerancia a Radiación/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...