Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Int J Pharm ; 606: 120846, 2021 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-34216769

RESUMO

Essential oils (EOs) of Thymus capitatus (Th) carvacrol chemotype and Origanum vulgare (Or) thymol and carvacrol chemotype were encapsulated in biocompatible poly(ε-caprolactone) nanocapsules (NCs). These nanosystems exhibited antibacterial, antifungal, and antibiofilm activities against Staphylococcus aureus, Escherichia coli, and Candida albicans. Th-NCs and Or-NCs were more effective against all tested strains than pure EOs and at the same time were not cytotoxic on HaCaT (T0020001) human keratinocyte cell line. The genotoxic effects of EO-NCs and EOs on HaCaT were evaluated using an alkaline comet assay for the first time, revealing that Th-NCs and Or-NCs did not induce DNA damage compared with untreated control HaCaT cells in vitro after 24 h. The cells morphological changes were assessed by label-free live cell Raman imaging. This study demonstrate the ability of poly(ε-caprolactone) nanocapsules loaded with thyme and oregano EOs to reduce microbial and biofilm growth and could be an ecological alternative in the development of new antimicrobial strategies.


Assuntos
Nanocápsulas , Óleos Voláteis , Antibacterianos/toxicidade , Biofilmes , Linhagem Celular , Dano ao DNA , Humanos , Queratinócitos , Testes de Sensibilidade Microbiana , Óleos Voláteis/farmacologia , Poliésteres
2.
Nanomedicine ; 13(1): 69-80, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27593490

RESUMO

Iron oxide nanoparticles are one of the most promising types of nanoparticles for biomedical applications, primarily in the context of nanomedicine-based diagnostics and therapy; hence, great attention should be paid to their bio-safety. Here, we investigate the ability of surface-modified magnetite nanoparticles (MNPs) to produce chromosome damage in human alveolar A549 cells. Compared to control cells, all the applied MNPs increased the level of micronuclei moderately but did not cause structural chromosomal aberrations in exposed cells. A rise in endoreplication, polyploid and multinuclear cells along with disruption of tubulin filaments, downregulation of Aurora protein kinases and p53 protein activation indicated the capacity of these MNPs to impair the chromosomal passenger complex and/or centrosome maturation. We suppose that surface-modified MNPs may act as aneugen-like spindle poisons via interference with tubulin polymerization. Further studies on experimental animals revealing mechanisms of therapeutic-aimed MNPs are required to confirm their suitability as potential anti-cancer drugs.


Assuntos
Aneugênicos/farmacologia , Antineoplásicos/farmacologia , Nanopartículas de Magnetita/química , Fuso Acromático/efeitos dos fármacos , Células A549 , Dano ao DNA , Humanos , Micronúcleos com Defeito Cromossômico , Nanomedicina , Tubulina (Proteína)/efeitos dos fármacos
3.
Toxicol Lett ; 226(3): 303-13, 2014 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-24614527

RESUMO

The generation of reactive oxygen species (ROS) has been proposed as the underlying mechanism involved in the genotoxicity of iron oxide nanoparticles. The data published to date are, however, inconsistent, and the mechanism underlying ROS formation has not been completely elucidated. Here, we investigated the capacity of several surface-modified magnetite nanoparticles (MNPs) to generate ROS in A549 human lung adenocarcinoma epithelial cells and HEL 12469 human embryonic lung fibroblasts. All MNPs, regardless of the coating, induced significant levels of DNA breakage in A549 cells but not in HEL 12469 cells. Under the same treatment conditions, variable low levels of intracellular ROS were detected in both A549 and HEL 12469 cells, but compared with control treatment, none of the coated MNPs produced any significant increase in oxidative damage to DNA in either of these cell lines. Indeed, no significant changes in the total antioxidant capacity and intracellular glutathione levels were observed in MNPs-treated human lung cell lines regardless of surface coating. In line with these results, none of the surface-modified MNPs increased significantly the GPx activity in A549 cells and the SOD activity in HEL 12469 cells. The GPx activity was significantly increased only in SO-Fe3O4-treated HEL 12469 cells. The SOD activity was significantly increased in SO-PEG-PLGA-Fe3O4-treated A549 cells but significantly decreased in SO-Fe3O4-treated A549 cells. Our data indicate that oxidative stress plays, at most, only a marginal role in the genotoxicity of surface-modified MNPs considered in this study in human lung cells.


Assuntos
Dano ao DNA , Pulmão/efeitos dos fármacos , Nanopartículas de Magnetita/toxicidade , Espécies Reativas de Oxigênio/metabolismo , Apoptose , Células Cultivadas , Glutationa/análise , Glutationa Peroxidase/metabolismo , Humanos , Pulmão/metabolismo , Superóxido Dismutase/metabolismo
4.
Mutat Res ; 743(1-2): 91-8, 2012 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-22306306

RESUMO

Differences between tissues in the expression of drug-metabolizing enzymes may substantially contribute to tissue-specificity of chemical carcinogens. To verify this hypothesis, the spontaneously immortalized human keratinocytes HaCaT were used, in order to evaluate the genotoxic potential of 7H-dibenzo[c,g]carbazole (DBC), a known hepatocarcinogen and sarcomagen, and its synthetic tissue-specific derivatives, 5,9-dimethyl-DBC (DiMeDBC) and N-methyl-DBC (N-MeDBC), which manifest specific tropism to the liver and skin, respectively. HaCaT cells mainly express cytochrome P4501A1 (CYP1A1), which is involved in metabolism of DBC and N-MeDBC, but not DiMeDBC [10]. Both DBC and the sarcomagen N-MeDBC induced significant levels of DNA strand-breaks, micronuclei, and DNA adducts followed by the phosphorylation of the p53 protein and histone H2AX in HaCaT cells. In contrast, the specific hepatocarcinogen DiMeDBC was devoid of any significant genotoxic activity in this cell line. Our study demonstrates that the absence of drug-metabolizing enzyme(s) involved in DiMeDBC metabolism may contribute substantially to the tissue-specific genotoxicity of this hepatocarcinogen.


Assuntos
Carbazóis/toxicidade , Carcinógenos/toxicidade , Queratinócitos/efeitos dos fármacos , Mutagênicos/toxicidade , Carbazóis/química , Linhagem Celular , Citocromo P-450 CYP1A1/metabolismo , Quebras de DNA de Cadeia Simples , Humanos , Queratinócitos/metabolismo , Testes de Mutagenicidade , Especificidade de Órgãos
5.
Environ Mol Mutagen ; 52(8): 636-45, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21809388

RESUMO

The goal of this study was to investigate the genotoxicity of 7H-dibenzo[c,g]carbazole (DBC), a ubiquitous environmental pollutant, and its methyl derivatives, 5,9-dimethylDBC (DiMeDBC), a strict hepatocarcinogen, and N-methylDBC (N-MeDBC), a specific sarcomagen in human hepatoma HepG2 cells, and to infer potential mechanisms underlying the biological activity of particular carcinogen. All dibenzocarbazoles, regardless the tissue specificity, induced significant DNA strand break levels and micronuclei in HepG2 cells; though a mitotic spindle dysfunction rather than a chromosome breakage was implicated in N-MeDBC-mediated micronucleus formation. While DBC and N-MeDBC produced stable DNA adducts followed with p53 protein phosphorylation at Ser-15, DiMeDBC failed. A significant increase in DNA strand breaks following incubation of exposed cells with a repair-specific endonuclease (Fpg protein) suggested that either oxidative DNA damage or unstable DNA-adducts might underlie DiMeDBC genotoxicity in human hepatoma cells. DiMeDBC and N-MeDBC increased substantially also the amount of CYP1A1/2 expression in HepG2 cells. Pretreatment of cells with substances affecting AhR-mediated CYP1A family of enzymes expression; however, diminished DiMeDBC and N-MeDBC genotoxicity. Our data clearly demonstrated differences in the mechanisms involved in the biological activity of DiMeDBC and N-MeDBC in human hepatoma cells; the genotoxicity of these DBC derivatives is closely related to CYP1A1/2 expression.


Assuntos
Carbazóis/toxicidade , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A2/genética , Mutagênicos/toxicidade , Sequência de Bases , Western Blotting , Sobrevivência Celular/efeitos dos fármacos , Ensaio Cometa , Adutos de DNA , Quebras de DNA , Relação Dose-Resposta a Droga , Células Hep G2 , Histonas/metabolismo , Humanos , Micronúcleos com Defeito Cromossômico/induzido quimicamente , Micronúcleos com Defeito Cromossômico/estatística & dados numéricos , Testes para Micronúcleos , Índice Mitótico , Dados de Sequência Molecular , Fosforilação , Reação em Cadeia da Polimerase em Tempo Real , Proteína Supressora de Tumor p53/metabolismo
6.
Toxicol Appl Pharmacol ; 255(3): 307-15, 2011 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-21798277

RESUMO

The environmental pollutant 7H-dibenzo[c,g]carbazole (DBC) and its derivative, 5,9-dimethylDBC (DiMeDBC), produced significant and dose-dependent levels of micronuclei followed by a substantial increase in the frequency of apoptotic cells in the V79MZh3A4 cell line stably expressing the human cytochrome P450 (hCYP) 3A4. In contrast, neither micronuclei nor apoptosis were found in cells exposed to the sarcomagenic carcinogen, N-methylDBC (N-MeDBC). A slight but significant level of gene mutations and DNA adducts detected in V79MZh3A4 cells treated with N-MeDBC, only at the highest concentration (30µM), revealed that this sarcomagenic carcinogen was also metabolized by hCYP3A4. Surprisingly, DBC increased the frequency of 6-thioguanine resistant (6-TG(r)) mutations only at the highest concentration (30µM), while DiMeDBC failed to increase the frequency of these mutations. The resistance to 6-thioguanine is caused by the mutations in the hypoxanthine-guanine phosphoribosyltransferase (Hprt) gene. The molecular analysis of the coding region of Hprt gene showed a deletion of the entire exon 8 in DiMeDBC-induced 6-TG(r) mutants, while no changes in the nucleotide sequences were identified in 6-TG(r) mutants produced by DBC and N-MeDBC. Based on our results, we suggest that hCYP3A4 is involved in the metabolism of DBC and its tissue-specific derivatives. While hCYP3A4 probably plays an important role in biotransformation of the liver carcinogens, DBC and DiMeDBC, it might only have a marginal function in N-MeDBC metabolism.


Assuntos
Carbazóis/metabolismo , Citocromo P-450 CYP3A/fisiologia , Animais , Biotransformação/efeitos dos fármacos , Biotransformação/genética , Linhagem Celular , Cricetinae , Cricetulus , Citocromo P-450 CYP3A/genética , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA