Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
1.
Nucleic Acids Res ; 48(W1): W455-W462, 2020 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-32421831

RESUMO

In the past few decades, major initiatives have been launched around the world to address chemical safety testing. These efforts aim to innovate and improve the efficacy of existing methods with the long-term goal of developing new risk assessment paradigms. The transcriptomic and toxicological profiling of mammalian cells has resulted in the creation of multiple toxicogenomic datasets and corresponding tools for analysis. To enable easy access and analysis of these valuable toxicogenomic data, we have developed ToxicoDB (toxicodb.ca), a free and open cloud-based platform integrating data from large in vitro toxicogenomic studies, including gene expression profiles of primary human and rat hepatocytes treated with 231 potential toxicants. To efficiently mine these complex toxicogenomic data, ToxicoDB provides users with harmonized chemical annotations, time- and dose-dependent plots of compounds across datasets, as well as the toxicity-related pathway analysis. The data in ToxicoDB have been generated using our open-source R package, ToxicoGx (github.com/bhklab/ToxicoGx). Altogether, ToxicoDB provides a streamlined process for mining highly organized, curated, and accessible toxicogenomic data that can be ultimately applied to preclinical toxicity studies and further our understanding of adverse outcomes.


Assuntos
Bases de Dados Genéticas , Software , Toxicogenética/métodos , Acetaminofen/toxicidade , Animais , Gráficos por Computador , DNA/biossíntese , Mineração de Dados , Expressão Gênica/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Inibidores da Síntese de Ácido Nucleico/toxicidade , Ratos
2.
Basic Clin Pharmacol Toxicol ; 122(6): 633-642, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29285878

RESUMO

Amanita phalloides species mushrooms containing alpha-amanitin (α-AMA) are responsible for the majority of fatal mushroom intoxications and can lead to severe poisonings resulting in hepatotoxicity and acute hepatic failure. Existing antidotes, such as silibinin, are not sufficiently effective in the prevention and/or resolution of α-AMA-induced hepatotoxicity. We investigated the effects of resveratrol on α-AMA-induced hepatotoxicity and compared with silibinin, a known antidote using in vivo and in vitro toxicity models. In the in vivo protocol, resveratrol (30 mg/kg) was given simultaneously with α-AMA (α-AMA + SR) or 12 (α-AMA + 12R) or 24 (α-AMA + 24R) hr after α-AMA administration. Silibinin (5 mg/kg) (α-AMA + Sil) and normal saline (α-AMA + NS) were given simultaneously with α-AMA. We found that liver transaminase levels in α-AMA + SR and α-AMA + 12R groups and histomorphologic injury score in the α-AMA + SR, α-AMA + 12R, α-AMA + 24R and α-AMA + Sil groups were significantly lower than that of the α-AMA + NS group. Resveratrol decreased mononuclear cell infiltration, necrosis and active caspase-3 immunopositivity in the liver. In the in vitro protocol, the effects of resveratrol and silibinin were evaluated in a reduction in cell viability induced by α-AMA in THLE-2 and THLE-3 hepatocytes. Neither resveratrol nor silibinin was found to be effective in increasing cell viability decreased by α-AMA + NS. As a conclusion, resveratrol was found to be effective in α-AMA-induced hepatotoxicity with its anti-inflammatory properties in in vivo conditions. It is a promising compound with the potential for use in the treatment of hepatotoxicity associated with Amanita phalloides type mushroom poisonings.


Assuntos
Alfa-Amanitina/antagonistas & inibidores , Alfa-Amanitina/toxicidade , Antioxidantes/uso terapêutico , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Intoxicação Alimentar por Cogumelos/tratamento farmacológico , Inibidores da Síntese de Ácido Nucleico/toxicidade , Substâncias Protetoras/uso terapêutico , Silimarina/uso terapêutico , Estilbenos/uso terapêutico , Alanina Transaminase/sangue , Aspartato Aminotransferases/sangue , Caspase 3/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Doença Hepática Induzida por Substâncias e Drogas/patologia , Humanos , Fígado/enzimologia , Fígado/patologia , Resveratrol , Silibina
3.
Food Chem Toxicol ; 101: 48-54, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28069374

RESUMO

The significant contents of artepillin C (AC) in green propolis have prompted research on the biological activities of the compound. The present study evaluated the activity of this phenolic compound on DNA, assessing its genotoxic and antigenotoxic potentials in the somatic mutation and recombination test in Drosophila melanogaster. The standard (ST) and high-bioactivation (HB) crosses were used in the assessment of genotoxic potential, since they express cytochrome P450 metabolization enzymes differently. In the 0.1-1.6 mM concentration range, AC did not have any genotoxic action in either cross. Antigenotoxic potential was investigated using the ST cross. In co- and post-treatment protocols, AC 0.4, 0.8, and 1.6 mM did not modulate mutagenic action of ethyl methanesulphonate. However, though it did not influence the frequency of damage induced by mitomycin C in co-treatment, AC reduced genotoxicity of the mutagen when administered after damage, but only at 0.4 mM. This modulation is associated with the reduction of genetic damage caused by recombinational events. The results of the present study and literature findings indicate that the various responses elicited by AC, namely induction of DNA damage, production of genetic lesions, or activation of DNA repair mechanisms are functions of AC concentration.


Assuntos
Dano ao DNA/efeitos dos fármacos , Drosophila melanogaster/efeitos dos fármacos , Fenilpropionatos/toxicidade , Recombinação Genética/genética , Animais , Células Cultivadas , Dano ao DNA/genética , Drosophila melanogaster/genética , Testes de Mutagenicidade/métodos , Inibidores da Síntese de Ácido Nucleico/toxicidade
4.
Genetics ; 204(3): 959-973, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27585850

RESUMO

Hydroxyurea (HU) has been used for the treatment of multiple diseases, such as cancer. The therapeutic effect is generally believed to be due to the suppression of ribonucleotide reductase (RNR), which slows DNA polymerase movement at replication forks and induces an S phase cell cycle arrest in proliferating cells. Although aberrant mitosis and DNA damage generated at collapsed forks are the likely causes of cell death in the mutants with defects in replication stress response, the mechanism underlying the cytotoxicity of HU in wild-type cells remains poorly understood. While screening for new fission yeast mutants that are sensitive to replication stress, we identified a novel mutation in the erg11 gene encoding the enzyme sterol-14α-demethylase in the ergosterol biosynthesis pathway that dramatically sensitizes the cells to chronic HU treatment. Surprisingly, HU mainly arrests the erg11 mutant cells in cytokinesis, not in S phase. Unlike the reversible S phase arrest in wild-type cells, the cytokinesis arrest induced by HU is relatively stable and occurs at low doses of the drug, which likely explains the remarkable sensitivity of the mutant to HU. We also show that the mutation causes sterol deficiency, which may predispose the cells to the cytokinesis arrest and lead to cell death. We hypothesize that in addition to the RNR, HU may have a secondary unknown target(s) inside cells. Identification of such a target(s) may greatly improve the chemotherapies that employ HU or help to expand the clinical usage of this drug for additional pathological conditions.


Assuntos
Citocinese , Ergosterol/biossíntese , Hidroxiureia/toxicidade , Inibidores da Síntese de Ácido Nucleico/toxicidade , Proteínas de Schizosaccharomyces pombe/genética , Schizosaccharomyces/metabolismo , Esterol 14-Desmetilase/genética , Mutação , Schizosaccharomyces/citologia , Schizosaccharomyces/efeitos dos fármacos , Schizosaccharomyces/genética , Proteínas de Schizosaccharomyces pombe/metabolismo , Esterol 14-Desmetilase/metabolismo
6.
Food Chem Toxicol ; 84: 250-9, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26363308

RESUMO

Microbial detoxification of deoxynivalenol (DON) represents a new approach to treating DON-contaminated grains. A bacterium Devosia mutans 17-2-E-8 was capable of completely transforming DON into a major product 3-epi-DON and a minor product 3-keto-DON. Evaluation of toxicities of these DON-transformation products is an important part of hazard characterization prior to commercialization of the biotransformation application. Cytotoxicities of the products were demonstrated by two assays: a MTT bioassay assessing cell viability and a BrdU assay assessing DNA synthesis. Compared with DON, the IC50 values of 3-epi-DON and 3-keto-DON were respectively 357 and 3.03 times higher in the MTT bioassay, and were respectively 1181 and 4.54 times higher in the BrdU bioassay. Toxicological effects of 14-day oral exposure of the B6C3F1 mouse to DON and 3-epi-DON were also investigated. Overall, there were no differences between the control (free of toxin) and the 25 mg/kg bw/day or 100 mg/kg bw/day 3-epi-DON treatments in body and organ weights, hematology and organ histopathology. However, in mice exposed to DON (2 mg/kg bw/day), white blood cell numbers and serum immunoglobulin levels were altered relative to controls, and lesions were observed in adrenals, thymus, stomach, spleen and colon. Taken together, in vitro and in vivo studies indicate that 3-epi-DON is substantially less toxic than DON.


Assuntos
Hyphomicrobiaceae/metabolismo , Inibidores da Síntese de Ácido Nucleico/toxicidade , Tricotecenos/toxicidade , Administração Oral , Animais , Células CACO-2 , Sobrevivência Celular/efeitos dos fármacos , Cruzamentos Genéticos , DNA/biossíntese , Relação Dose-Resposta a Droga , Feminino , Humanos , Inativação Metabólica , Cinética , Camundongos , Células NIH 3T3 , Inibidores da Síntese de Ácido Nucleico/administração & dosagem , Inibidores da Síntese de Ácido Nucleico/química , Inibidores da Síntese de Ácido Nucleico/metabolismo , Distribuição Aleatória , Estereoisomerismo , Testes de Toxicidade Subaguda , Tricotecenos/administração & dosagem , Tricotecenos/química , Tricotecenos/metabolismo
7.
Brain Res Bull ; 109: 151-7, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25451454

RESUMO

To identify an individual as familiar, rodents form a specific type of memory named social recognition memory. The olfactory bulb (OB) is an important structure for social recognition memory, while the hippocampus recruitment is still controversial. The present study was designed to elucidate the OB and the dorsal hippocampus contribution to the consolidation of social memory. For that purpose, we tested the effect of anisomycin (ANI), which one of the effects is the inhibition of protein synthesis, on the consolidation of social recognition memory. Swiss adult mice with cannulae implanted into the CA1 region of the dorsal hippocampus or into the OB were exposed to a juvenile during 5 min (training session; TR), and once again 1.5 h or 24 h later to test social short-term memory (S-STM) or social long-term memory (S-LTM), respectively. To study S-LTM consolidation, mice received intra-OB or intra-CA1 infusion of saline or ANI immediately, 3, 6 or 18 h after TR. ANI impaired S-LTM consolidation in the OB, when administered immediately or 6h after TR. In the dorsal hippocampus, ANI was amnesic only if administered 3 h after TR. Furthermore, the infusion of ANI in either OB or CA1, immediately after training, did not affect S-STM. Moreover, ANI administered into the OB did not alter the animal's performance in the buried food-finding task. Altogether, our results suggest the consolidation of S-LTM requires both OB and hippocampus participation, although in different time points. This study may help shedding light on the specific roles of the OB and dorsal hippocampus in social recognition memory.


Assuntos
Anisomicina/toxicidade , Hipocampo/efeitos dos fármacos , Transtornos da Memória/induzido quimicamente , Inibidores da Síntese de Ácido Nucleico/toxicidade , Bulbo Olfatório/efeitos dos fármacos , Reconhecimento Psicológico/efeitos dos fármacos , Comportamento Social , Fatores Etários , Animais , Condicionamento Psicológico/efeitos dos fármacos , Modelos Animais de Doenças , Medo/psicologia , Comportamento Alimentar/efeitos dos fármacos , Masculino , Camundongos , Tempo de Reação/efeitos dos fármacos , Estatísticas não Paramétricas , Fatores de Tempo
8.
Biochem Biophys Res Commun ; 447(4): 586-9, 2014 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-24747075

RESUMO

The Marasmius oreades mushroom agglutinin (MOA) is a blood group B-specific lectin carrying an active proteolytic domain. Its enzymatic activity has recently been shown to be critical for toxicity of MOA toward the fungivorous soil nematode Caenorhabditis elegans. Here we present evidence that MOA also induces cytotoxicity in a cellular model system (murine NIH/3T3 cells), by inhibiting protein synthesis, and that cytotoxicity correlates, at least in part, with proteolytic activity. A peptide-array screen identified the apoptosis mediator BAX as a potential proteolytic substrate and further suggests a variety of bacterial and fungal peptides as potential substrates. These findings are in line with the suggestion that MOA and related proteases may play a role for host defense.


Assuntos
Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Proteínas Fúngicas/farmacologia , Proteína X Associada a bcl-2/metabolismo , Aglutininas/metabolismo , Aglutininas/farmacologia , Aglutininas/toxicidade , Substituição de Aminoácidos , Animais , Proteínas Fúngicas/metabolismo , Proteínas Fúngicas/toxicidade , Variação Genética , Lectinas/metabolismo , Lectinas/farmacologia , Lectinas/toxicidade , Marasmius/química , Marasmius/genética , Camundongos , Células NIH 3T3 , Inibidores da Síntese de Ácido Nucleico/farmacologia , Inibidores da Síntese de Ácido Nucleico/toxicidade , Inibidores da Síntese de Proteínas/farmacologia , Inibidores da Síntese de Proteínas/toxicidade
9.
Toxicol Lett ; 224(3): 395-406, 2014 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-24247028

RESUMO

Previously, we studied the effects of deoxynivalenol (DON) and tributyltin oxide (TBTO) on whole genome mRNA expression profiles of human T lymphocyte Jurkat cells. These studies indicated that DON induces ribotoxic stress and both DON and TBTO induced ER stress which resulted into T-cell activation and apoptosis. The first goal of the present study was to provide final proof for these mode of actions by comparing the effects of 6 h exposure to DON and TBTO on mRNA expression to those of positive controls of ribotoxic stress (anisomycin), ER stress (thapsigargin) and T cell activation (ionomycin). Genes affected by anisomycin and the majority of genes affected by thapsigargin were affected in the same direction by DON and TBTO, respectively, confirming the expected modes of action. Pathway analysis further sustained that DON induces ribotoxic stress and both DON and TBTO induce unfolded protein response (UPR), ER stress, T cell activation and apoptosis. The second goal was to assess whether DON and/or TBTO affect other pathways above those detected before. TBTO induced groups of genes that are involved in DNA packaging and heat shock response that were not affected by thapsigargin. DON did not affect other genes than anisomycin indicating the effect of DON to be restricted to ribotoxic stress. This study also demonstrates that comparative gene expression analysis is a very promising tool for the identification of modes of action of immunotoxic compounds.


Assuntos
Anisomicina/toxicidade , Carcinógenos/toxicidade , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Perfilação da Expressão Gênica , Inibidores da Síntese de Ácido Nucleico/toxicidade , Tapsigargina/toxicidade , Compostos de Trialquitina/toxicidade , Tricotecenos/toxicidade , Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Mapeamento Cromossômico , Interpretação Estatística de Dados , Proteínas de Choque Térmico/metabolismo , Humanos , Ionomicina/farmacologia , Células Jurkat , Análise em Microsséries , Proteínas Mitocondriais/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , RNA Neoplásico/biossíntese , RNA Neoplásico/isolamento & purificação , Linfócitos T/efeitos dos fármacos
10.
Toxicol Sci ; 133(2): 298-308, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23492809

RESUMO

Hydroxyurea (HU) is commonly used to treat myeloproliferative diseases and sickle cell anemia. The administration of HU to gestation day 9 CD1 mice causes predominantly hindlimb, tail, and neural tube defects. HU induces oxidative stress and p38 mitogen-activated protein kinase (MAPK) signaling in embryos. HU also inactivates ribonucleotide reductase, leading to DNA replication stress and DNA damage response signaling. We hypothesize that HU exposure induces p38 MAPK activation and DNA damage response signaling during organogenesis preferentially in malformation-sensitive tissues. HU treatment (400 or 600mg/kg) induced the activation of MEK3/6, upstream MAP2K3 kinases, within 30min; phospho-MEK3/6 immunoreactivity was increased throughout the embryo. Activation of the downstream p38 MAPK peaked 3h post-HU treatment. At this time, phospho-p38 MAPK immunoreactivity was enhanced in the cytoplasm and nucleus of cells in the rostral and caudal neuroepithelium and neural tube; significant increases in p38 MAPK signaling were not observed in the somites or heart. Interestingly, the DNA damage response, as assessed by the formation of γH2AX foci, was increased at 3h in HU-exposed embryos in all tissues examined, including the somites and heart. Increases in pyknotic nuclei and cell fragmentation were observed in all tissues except the heart, an organ that is relatively resistant to HU-induced malformations. Thus, although HU induces a widespread DNA damage response, the activation of p38 MAPK is localized to the rostral and caudal neuroepithelium and neural tube, suggesting that p38 MAPK pathways may play a role in mediating the specific malformations observed after HU exposure.


Assuntos
DNA/efeitos dos fármacos , Embrião de Mamíferos/efeitos dos fármacos , Hidroxiureia/toxicidade , Inibidores da Síntese de Ácido Nucleico/toxicidade , Transdução de Sinais/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Anormalidades Induzidas por Medicamentos , Animais , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Citoplasma/efeitos dos fármacos , Citoplasma/metabolismo , Dano ao DNA , Relação Dose-Resposta a Droga , Técnicas de Cultura Embrionária , Embrião de Mamíferos/metabolismo , Feminino , MAP Quinase Quinase 3/metabolismo , MAP Quinase Quinase 6/metabolismo , Camundongos , Camundongos Endogâmicos , Necrose/induzido quimicamente , Fosforilação , Gravidez
11.
Toxicol Appl Pharmacol ; 265(1): 10-8, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-23022514

RESUMO

The Type B trichothecene deoxynivalenol (DON), a ribotoxic mycotoxin known to contaminate cereal-based foods, induces ribosomal RNA (rRNA) cleavage in the macrophage via p38-directed activation of caspases. Here we employed the RAW 264.7 murine macrophage model to test the hypothesis that this rRNA cleavage pathway is similarly induced by other ribotoxins. Capillary electrophoresis confirmed that the antibiotic anisomycin (≥25ng/ml), the macrocylic trichothecene satratoxin G (SG) (≥10ng/ml) and ribosome-inactivating protein ricin (≥300ng/ml) induced 18s and 28s rRNA fragmentation patterns identical to that observed for DON. Also, as found for DON, inhibition of p38, double-stranded RNA-activated kinase (PKR) and hematopoietic cell kinase (Hck) suppressed MAPK anisomycin-induced rRNA cleavage, while, in contrast, their inhibition did not affect SG- and ricin-induced rRNA fragmentation. The p53 inhibitor pifithrin-µ and pan caspase inhibitor Z-VAD-FMK suppressed rRNA cleavage induced by anisomycin, SG and ricin, indicating that these ribotoxins shared with DON a conserved downstream pathway. Activation of caspases 8, 9 and 3 concurrently with apoptosis further suggested that rRNA cleavage occurred in parallel with both extrinsic and intrinsic pathways of programmed cell death. When specific inhibitors of cathepsins L and B (lysosomal cysteine cathepsins active at cytosolic neutral pH) were tested, only the former impaired anisomycin-, SG-, ricin- and DON-induced rRNA cleavage. Taken together, the data suggest that (1) all four ribotoxins induced p53-dependent rRNA cleavage via activation of cathepsin L and caspase 3, and (2) activation of p53 by DON and anisomycin involved p38 whereas SG and ricin activated p53 by an alternative mechanism.


Assuntos
Clivagem do RNA/efeitos dos fármacos , RNA Ribossômico/efeitos dos fármacos , Tricotecenos/toxicidade , Animais , Anisomicina/toxicidade , Apoptose/efeitos dos fármacos , Western Blotting , Caspase 8/efeitos dos fármacos , Catepsina L/farmacologia , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Inibidores da Síntese de Ácido Nucleico/toxicidade , Proteínas Proto-Oncogênicas c-hck/metabolismo , RNA Ribossômico/isolamento & purificação , Ricina/toxicidade , Tricotecenos/farmacologia , Proteína Supressora de Tumor p53/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
12.
J Nutr ; 142(11): 1983-90, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23054309

RESUMO

It is unknown what feeding strategy to use during chemotherapy-induced gastrointestinal mucositis, which causes weight loss and possibly malabsorption. To study the absorptive capacity of amino acids during mucositis, we determined the plasma availability of enterally administered amino acids (AA), their utilization for protein synthesis, and the preferential side of the intestine for AA uptake in rats with and without methotrexate (MTX)-induced mucositis. Four days after injection with MTX (60 mg/kg) or saline (controls), rats received a primed, continuous dual-isotope infusion (intraduodenal and intravenous) of labeled L-leucine, L-lysine, L-phenylalanine, L-threonine, and L-methionine. We collected blood samples, assessed jejunal histology, and determined labeled AA incorporation in proximal and distal small intestinal mucosa, plasma albumin, liver, and thigh muscle. MTX-induced mucositis was confirmed by histology. The median systemic availability of all AA except for leucine was similar in MTX-treated rats and in controls. However, the individual availability of all AA differed substantially within the group of MTX-treated rats, ranging from severely reduced (<10% of intake) to not different from controls (>40% of intake in 5 of 9 rats). More AA originating from basolateral uptake than those originating from apical uptake were used for intestinal protein synthesis in MTX-treated rats (≥420% more, P < 0.05). We conclude that continuous enteral administration can enable normal AA absorption in rats with MTX-induced mucositis. The intestine prefers basolateral AA uptake to meet its need for AA for protein synthesis during mucositis.


Assuntos
Aminoácidos/metabolismo , Metotrexato/toxicidade , Mucosite/induzido quimicamente , Inibidores da Síntese de Ácido Nucleico/toxicidade , Absorção , Albuminas/metabolismo , Animais , Nutrição Enteral , Regulação da Expressão Gênica , Injeções Intravenosas , Mucosa Intestinal/metabolismo , Masculino , Músculo Esquelético/metabolismo , Ratos , Ratos Wistar , Organismos Livres de Patógenos Específicos
13.
J Toxicol Sci ; 36(6): 695-712, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22129734

RESUMO

It is not widely known how the developing brain responds to extrinsic damage, although the developing brain is considered to be sensitive to diverse environmental factors including DNA-damaging agents. This paper reviews the mechanisms of neurotoxicity induced in the developing brain of mice and rats by six chemicals (ethylnitrosourea, hydroxyurea, 5-azacytidine, cytosine arabinoside, 6-mercaptopurine and etoposide), which cause DNA damage in different ways, especially from the viewpoints of apoptosis and cell cycle arrest in neural progenitor cells. In addition, this paper also reviews the repair process following damage in the developing brain.


Assuntos
Alquilantes/toxicidade , Dano ao DNA , Mutagênicos/toxicidade , Células-Tronco Neurais/efeitos dos fármacos , Inibidores da Síntese de Ácido Nucleico/toxicidade , Animais , Apoptose/efeitos dos fármacos , Encéfalo/citologia , Encéfalo/embriologia , Encéfalo/metabolismo , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Humanos , Camundongos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/metabolismo , Ratos
14.
Genetics ; 189(2): 533-47, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21840863

RESUMO

In Saccharomyces cerevisiae, replication stress induced by hydroxyurea (HU) and methyl methanesulfonate (MMS) activates DNA integrity checkpoints; in checkpoint-defective yeast strains, HU treatment also induces morphological aberrations. We find that the sphingolipid pathway gene ISC1, the product of which catalyzes the generation of bioactive ceramides from complex sphingolipids, plays a novel role in determining cellular morphology following HU/MMS treatment. HU-treated isc1Δ cells display morphological aberrations, cell-wall defects, and defects in actin depolymerization. Swe1, a morphogenesis checkpoint regulator, and the cell cycle regulator Cdk1 play key roles in these morphological defects of isc1Δ cells. A genetic approach reveals that ISC1 interacts with other checkpoint proteins to control cell morphology. That is, yeast carrying deletions of both ISC1 and a replication checkpoint mediator gene including MRC1, TOF1, or CSM3 display basal morphological defects, which increase following HU treatment. Interestingly, strains with deletions of both ISC1 and the DNA damage checkpoint mediator gene RAD9 display reduced morphological aberrations irrespective of HU treatment, suggesting a role for RAD9 in determining the morphology of isc1Δ cells. Mechanistically, the checkpoint regulator Rad53 partially influences isc1Δ cell morphology in a dosage-dependent manner.


Assuntos
Dano ao DNA , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/genética , Fosfolipases Tipo C/genética , Actinas/metabolismo , Western Blotting , Proteína Quinase CDC28 de Saccharomyces cerevisiae/genética , Proteína Quinase CDC28 de Saccharomyces cerevisiae/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Parede Celular/efeitos dos fármacos , Parede Celular/metabolismo , Quitina/metabolismo , Reparo do DNA , Replicação do DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Hidroxiureia/toxicidade , Metanossulfonato de Metila/toxicidade , Microscopia de Fluorescência , Mutagênicos/toxicidade , Mutação , Inibidores da Síntese de Ácido Nucleico/toxicidade , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/crescimento & desenvolvimento , Proteínas de Saccharomyces cerevisiae/metabolismo , Esfingolipídeos/metabolismo , Fosfolipases Tipo C/metabolismo
15.
Hum Exp Toxicol ; 30(1): 38-43, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20378659

RESUMO

α-Amanitin (α-AMA) is the main toxin of Amanita phalloides and its subspecies (A. virosa and A. verna). The primary mechanism of α-AMA toxicity is associated with protein synthesis blocking in hepatocytes. Additionally, α-AMA exhibits prooxidant properties that may contribute to its severe hepatotoxicity. The aim of the present study was to assess the effect of α-AMA on lipid peroxidation and the activities of superoxide dismutase (SOD) and catalase (CAT) in human hepatocyte culture. The effects of benzylpenicillin (BPCN), N-acetyl-L-cysteine (ACC), and silibinin (SIL) on SOD and CAT activities and on lipid peroxidation in human hepatocyte culture intoxicated with α-AMA were also examined. In human hepatocyte culture, 48-hour exposure to α-AMA at a 2-µM concentration caused an increase in SOD activity, a reduction of CAT activity, and a significant increase in lipid peroxidation. Changes in SOD and CAT activity caused by α-AMA could probably enhance lipid peroxidation by increased generation of hydrogen peroxide combined with reduced detoxification of that oxygen radical. The addition of antidotes (ACC or SIL) to the culture medium provided more effective protection against lipid peroxidation in human hepatocytes intoxicated with α-AMA than the addition of BPCN, possessing no antioxidant properties.


Assuntos
Alfa-Amanitina/toxicidade , Antídotos/farmacologia , Antioxidantes/farmacologia , Hepatócitos/efeitos dos fármacos , Inibidores da Síntese de Ácido Nucleico/toxicidade , Oxidantes/toxicidade , Acetilcisteína/farmacologia , Amanita/química , Catalase/metabolismo , Células Cultivadas , Hepatócitos/enzimologia , Hepatócitos/metabolismo , Humanos , Peroxidação de Lipídeos/efeitos dos fármacos , Intoxicação Alimentar por Cogumelos/tratamento farmacológico , Penicilina G/farmacologia , Silibina , Silimarina/farmacologia , Superóxido Dismutase/metabolismo
16.
Toxicol In Vitro ; 24(3): 707-12, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20123012

RESUMO

The chromosomal aberrations (CA) assay and micronucleus (MN) test were employed to investigate the effect in vitro of zerumbone (ZER) on human chromosomes. ZER is a sesquiterpene compound isolated from the rhizomes of wild ginger, Zingiber zerumbet Smith. The rhizomes of the plant are employed as a traditional medicine for some ailments and as condiments. ZER has been shown to have anti-cancer and apoptosis-inducing properties against various human tumour cells. It has also been shown to be active in vivo against a number of induced malignancies. Studies on ZER genotoxicity in cultured human peripheral blood lymphocytes (PBL) have not been reported so far. Therefore, the present study was undertaken to investigate the ability of ZER to induce chromosomal aberrations and micronuclei formation in human lymphocytes in vitro. Human blood samples were obtained from four healthy, non-smoking males aged 25-35years. Cultures were exposed to the drug for 48h at four final concentrations: 10, 20, 40 and 80 microM. Mitomycin C (MMC) was used as a positive control. The results of chromosomal aberrations assay showed that ZER was not clastogenic, when compared to untreated control, meanwhile MN test results showed a dose-dependent increase in MN formation. The overall clastogenic effect of ZER on human PBL was statistically not significant. In conclusion, ZER is a cytotoxic but not a clastogenic substance in human PBL.


Assuntos
Linfócitos/efeitos dos fármacos , Mutagênicos , Sesquiterpenos/toxicidade , Adulto , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Aberrações Cromossômicas/efeitos dos fármacos , Citocalasina B/antagonistas & inibidores , Citocalasina B/toxicidade , Zingiber officinale/química , Humanos , Masculino , Testes para Micronúcleos , Mitomicina/toxicidade , Índice Mitótico , Testes de Mutagenicidade , Inibidores da Síntese de Ácido Nucleico/toxicidade , Raízes de Plantas/química , Sesquiterpenos/isolamento & purificação , Adulto Jovem
17.
Antimicrob Agents Chemother ; 54(5): 1693-9, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20176897

RESUMO

By choosing membranes as targets of action, antibacterial peptides offer the promise of providing antibiotics to which bacteria would not become resistant. However, there is a need to increase their potency against bacteria along with achieving a reduction in toxicity to host cells. Here, we report that three de novo-designed antibacterial peptides (DeltaFm, DeltaFmscr, and Ud) with poor to moderate antibacterial potencies and kill kinetics improved significantly in all of these aspects when synergized with rifampin and kanamycin against Escherichia coli. (DeltaFm and DeltaFmscr [a scrambled-sequence version of DeltaFm] are isomeric, monomeric decapeptides containing the nonproteinogenic amino acid alpha,beta-didehydrophenylalanine [DeltaF] in their sequences. Ud is a lysine-branched dimeric peptide containing the helicogenic amino acid alpha-aminoisobutyric acid [Aib].) In synergy with rifampin, the MIC of DeltaFmscr showed a 34-fold decrease (67.9 microg/ml alone, compared to 2 microg/ml in combination). A 20-fold improvement in the minimum bactericidal concentration of Ud was observed when the peptide was used in combination with rifampin (369.9 microg/ml alone, compared to 18.5 microg/ml in combination). Synergy with kanamycin resulted in an enhancement in kill kinetics for DeltaFmscr (no killing until 60 min for DeltaFmscr alone, versus 50% and 90% killing within 20 min and 60 min, respectively, in combination with kanamycin). Combination of the dendrimeric peptide DeltaFq (a K-K2 dendrimer for which the sequence of DeltaFm constitutes each of the four branches) (MIC, 21.3 microg/ml) with kanamycin (MIC, 2.1 microg/ml) not only lowered the MIC of each by 4-fold but also improved the therapeutic potential of this highly hemolytic (37% hemolysis alone, compared to 4% hemolysis in combination) and cytotoxic (70% toxicity at 10x MIC alone, versus 30% toxicity in combination) peptide. Thus, synergy between peptide and nonpeptide antibiotics has the potential to enhance the potency and target selectivity of antibacterial peptides, providing regimens which are more potent, faster acting, and safer for clinical use.


Assuntos
Peptídeos Catiônicos Antimicrobianos/farmacologia , Infecções por Escherichia coli/tratamento farmacológico , Escherichia coli/efeitos dos fármacos , Canamicina/farmacologia , Rifampina/farmacologia , Animais , Antibacterianos/química , Antibacterianos/farmacologia , Antibacterianos/toxicidade , Peptídeos Catiônicos Antimicrobianos/toxicidade , Desenho de Fármacos , Sinergismo Farmacológico , Escherichia coli/crescimento & desenvolvimento , Fibroblastos/citologia , Células HeLa , Humanos , Canamicina/química , Canamicina/toxicidade , Camundongos , Testes de Sensibilidade Microbiana , Inibidores da Síntese de Ácido Nucleico/química , Inibidores da Síntese de Ácido Nucleico/farmacologia , Inibidores da Síntese de Ácido Nucleico/toxicidade , Rifampina/química , Rifampina/toxicidade
18.
Toxicol Appl Pharmacol ; 244(3): 263-72, 2010 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-20083129

RESUMO

PURPOSE: Environmental exposure to arsenic is an important public health issue. The effects of arsenic on different tissues and organs have been intensively studied. However, the effects of arsenic on bone marrow mesenchymal stem cells (MSCs) have not been reported. This study is designed to investigate the cell death process caused by arsenite and its related underlying mechanisms on MSCs. The rationale is that absorbed arsenic in the blood circulation can reach to the bone marrow and may affect the cell survival of MSCs. METHODS: MSCs of passage 1 were purchased from Tulane University, grown till 70% confluency level and plated according to the experimental requirements followed by treatment with arsenite at various concentrations and time points. Arsenite (iAs(III)) induced cytotoxic effects were confirmed by cell viability and cell cycle analysis. For the presence of canonic apoptosis markers; DNA damage, exposure of intramembrane phosphotidylserine, protein and m-RNA expression levels were analyzed. RESULTS: iAs(III) induced growth inhibition, G2-M arrest and apoptotic cell death in MSCs, the apoptosis induced by iAs(III) in the cultured MSCs was, via altering Bcl-2 family proteins and by involving intrinsic pathway. CONCLUSION: iAs(III) can induce apoptosis in bone marrow-derived MSCs via Bcl-2 family proteins, regulating intrinsic apoptotic pathway. Due to the multipotency of MSC, acting as progenitor cells for a variety of connective tissues including bone, adipose, cartilage and muscle, these effects of arsenic may be important in assessing the health risk of the arsenic compounds and understanding the mechanisms of arsenic-induced harmful effects.


Assuntos
Apoptose/efeitos dos fármacos , Arsenitos/toxicidade , Poluentes Ambientais/toxicidade , Células-Tronco Mesenquimais/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/efeitos dos fármacos , Caspase 3/metabolismo , Caspase 9/metabolismo , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Humanos , Células-Tronco Mesenquimais/metabolismo , Redes e Vias Metabólicas , Inibidores da Síntese de Ácido Nucleico/toxicidade , Poli(ADP-Ribose) Polimerases/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA Mensageiro/metabolismo , Regulação para Cima/efeitos dos fármacos , Proteína X Associada a bcl-2/metabolismo
19.
Indian J Exp Biol ; 48(8): 822-9, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21341541

RESUMO

Hepatoprotective activity of hydroalcoholic extract of Luffa acutangula (HAELA) against carbon tetrachloride (CCl4) and rifampicin-induced hepatotoxicity in rats was evaluated and probable mechanism(s) of action has been suggested. Administration of standard drug- silymarin and HAELA showed significant hepatoprotection against CCl4 and rifampicin induced hepatotoxicity in rats. Hepatoprotective activity of HAELA was due to the decreased levels of serum marker enzymes viz., (AST, ALT, ALP and LDH) and increased total protein including the improvement in histoarchitecture of liver cells of the treated groups as compared to the control group. HAELA also showed significant decrease in malondialdehyde (MDA) formation, increased activity of non-enzymatic intracellular antioxidant, glutathione and enzymatic antioxidants, catalase and superoxide dismutase. Results of this study demonstrated that endogenous antioxidants and inhibition of lipid peroxidation of membrane contribute to hepatoprotective activity of HAELA.


Assuntos
Tetracloreto de Carbono/toxicidade , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Luffa/química , Inibidores da Síntese de Ácido Nucleico/toxicidade , Extratos Vegetais/farmacologia , Rifampina/toxicidade , Administração Oral , Animais , Antioxidantes/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/patologia , Feminino , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Malondialdeído/metabolismo , Extratos Vegetais/administração & dosagem , Ratos , Ratos Wistar
20.
BMC Biol ; 7: 35, 2009 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-19558638

RESUMO

BACKGROUND: The p53 tumor suppressor and its related protein, p73, share a homologous DNA binding domain, and mouse genetics studies have suggested that they have overlapping as well as distinct biological functions. Both p53 and p73 are activated by genotoxic stress to regulate an array of cellular responses. Previous studies have suggested that p53 and p73 independently activate the cellular apoptotic program in response to cytotoxic drugs. The goal of this study was to compare the promoter-binding activity of p53 and p73 at steady state and after genotoxic stress induced by hydroxyurea. RESULTS: We employed chromatin immunoprecipitation, the NimbleGen promoter arrays and a model-based algorithm for promoter arrays to identify promoter sequences enriched in anti-p53 or anti-p73 immunoprecipitates, either before or after treatment with hydroxyurea, which increased the expression of both p53 and p73 in the human colon cancer cell line HCT116-3(6). We calculated a model-based algorithm for promoter array score for each promoter and found a significant correlation between the promoter occupancy profiles of p53 and p73. We also found that after hydroxyurea treatment, the p53-bound promoters were still bound by p73, but p73 became associated with additional promoters that that did not bind p53. In particular, we showed that hydroxyurea induces the binding of p73 but not p53 to the promoter of MLH3, which encodes a mismatch repair protein, and causes an up-regulation of the MLH3 mRNA. CONCLUSION: These results suggest that hydroxyurea exerts differential effects on the promoter-binding functions of p53 and p73 and illustrate the power of model-based algorithm for promoter array in the analyses of promoter occupancy profiles of highly homologous transcription factors.


Assuntos
Dano ao DNA , Proteínas de Ligação a DNA/metabolismo , Hidroxiureia/toxicidade , Proteínas Nucleares/metabolismo , Inibidores da Síntese de Ácido Nucleico/toxicidade , Regiões Promotoras Genéticas , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Algoritmos , Proteínas de Transporte/genética , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Técnicas de Transferência de Genes , Células HCT116 , Humanos , Hidroxiureia/metabolismo , Proteínas de Membrana/metabolismo , Modelos Biológicos , Proteínas MutL , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Inibidores da Síntese de Ácido Nucleico/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Fatores de Terminação de Peptídeos/genética , Ligação Proteica/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno , Proteína Tumoral p73 , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/genética , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA