Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 11(3): e0150219, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26950072

RESUMO

The chemoprotective properties of sulforaphane (SF), derived from cruciferous vegetables, are widely acknowledged to arise from its potent induction of xenobiotic-metabolizing and antioxidant enzymes. However, much less is known about the impact of SF on the efficacy of cancer therapy through the modulation of drug-metabolizing enzymes. To identify proteins modulated by a low concentration of SF, we treated HT29 colon cancer cells with 2.5 µM SF. Protein abundance changes were detected by stable isotope labeling of amino acids in cell culture. Among 18 proteins found to be significantly up-regulated, aldo-keto reductase 1C3 (AKR1C3), bioactivating the DNA cross-linking prodrug PR-104A, was further characterized. Preconditioning HT29 cells with SF reduced the EC50 of PR-104A 3.6-fold. The increase in PR-104A cytotoxicity was linked to AKR1C3 abundance and activity, both induced by SF in a dose-dependent manner. This effect was reproducible in a second colon cancer cell line, SW620, but not in other colon cancer cell lines where AKR1C3 abundance and activity were absent or barely detectable and could not be induced by SF. Interestingly, SF had no significant influence on PR-104A cytotoxicity in non-cancerous, immortalized human colonic epithelial cell lines expressing either low or high levels of AKR1C3. In conclusion, the enhanced response of PR-104A after preconditioning with SF was apparent only in cancer cells provided that AKR1C3 is expressed, while its expression in non-cancerous cells did not elicit such a response. Therefore, a subset of cancers may be susceptible to combined food-derived component and prodrug treatments with no harm to normal tissues.


Assuntos
Antineoplásicos/farmacologia , Neoplasias do Colo/patologia , Isotiocianatos/farmacologia , Compostos de Mostarda Nitrogenada/farmacologia , 3-Hidroxiesteroide Desidrogenases/metabolismo , Membro C3 da Família 1 de alfa-Ceto Redutase , Antineoplásicos/efeitos adversos , Transporte Biológico , Linhagem Celular Tumoral , Diploide , Sinergismo Farmacológico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Hidroxiprostaglandina Desidrogenases/metabolismo , Mucosa Intestinal/citologia , Mucosa Intestinal/efeitos dos fármacos , Isotiocianatos/metabolismo , Compostos de Mostarda Nitrogenada/efeitos adversos , Pró-Fármacos/efeitos adversos , Pró-Fármacos/farmacologia , Sulfóxidos
2.
J Biochem Mol Toxicol ; 29(1): 10-20, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25179160

RESUMO

Cellular induction of reductase enzymes can alter the susceptibility of cells toward drugs and chemicals. In this study, we compared the capacity of a single dose of sodium selenite and 3H-1,2-dithiole-3-thione (D3T) to influence the drug-relevant reducing capacity of HT29 cells over time, and defined the protein-specific contribution to this activity on the basis of selected reaction monitoring mass spectrometry. Thioredoxin reductase 1 (TrxR1) protein levels and activity were inducible up to 2.2-fold by selenium. In contrast, selenium had only a minor influence on prostaglandin reductase 1 (PTGR1) and NAD(P)H: quinone oxidoreductase 1 (NQO1) activity and protein levels. D3T, a strong Nrf2 inducer, induced all the reductases and additionally increased the cytotoxicity of hydroxymethylacylfulvene, a bioreductive DNA-alkylating drug. The data and experimental approaches allow one to define induction potency for reductase enzymes PTGR1, TrxR1, and NQO1 in HT29 cells and link these to changes in drug cytotoxicity.


Assuntos
Antineoplásicos/farmacologia , Neoplasias do Colo/enzimologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , NAD(P)H Desidrogenase (Quinona)/biossíntese , Proteínas de Neoplasias/metabolismo , Selenito de Sódio/farmacologia , Tionas/farmacologia , Tiofenos/farmacologia , Tiorredoxina Redutase 1/biossíntese , Oligoelementos/farmacologia , Linhagem Celular Tumoral , Neoplasias do Colo/tratamento farmacológico , Indução Enzimática/efeitos dos fármacos , Humanos , Fator 2 Relacionado a NF-E2/metabolismo
3.
Chem Res Toxicol ; 27(3): 377-86, 2014 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-24383545

RESUMO

Diets enriched with bioactive food components trigger molecular changes in cells that may contribute to either health-promoting or adverse effects. Recent technological advances in high-throughput data generation allow for observing systems-wide molecular responses to cellular perturbations with nontoxic and dietary-relevant doses while considering the intrinsic differences between cancerous and noncancerous cells. In this chemical profile, we compared molecular responses of the colon cancer cell line HT29 and a noncancerous colon epithelial cell line (HCEC) to two widely encountered food components, sulforaphane and selenium. We conducted this comparison by generating new transcriptome data by microarray gene-expression profiling, analyzing them statistically on the single gene, network, and functional pathway levels, and integrating them with protein expression data. Sulforaphane and selenium, at doses that did not inhibit the growth of the tested cells, induced or repressed the transcription of a limited number of genes in a manner distinctly dependent on the chemical and the cell type. The genes that most strongly responded in cancer cells were observed after treatment with sulforaphane and were members of the aldo-keto reductase (AKR) superfamily. These genes were in high agreement in terms of fold change with their corresponding proteins (correlation coefficient r(2) = 0.98, p = 0.01). Conversely, selenium had little influence on the cancer cells. In contrast, in noncancerous cells, selenium induced numerous genes involved in apoptotic, angiogenic, or tumor proliferation pathways, whereas the influence of sulforaphane was very limited. These findings contribute to defining the significance of cell type in interpreting human cellular transcriptome-level responses to exposures to natural components of the diet.


Assuntos
Isotiocianatos/toxicidade , Selênio/toxicidade , Transcriptoma/efeitos dos fármacos , 20-Hidroxiesteroide Desidrogenases/genética , 20-Hidroxiesteroide Desidrogenases/metabolismo , Linhagem Celular , Colo/citologia , Regulação para Baixo/efeitos dos fármacos , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células HT29 , Humanos , Isotiocianatos/química , Marcação por Isótopo , Redes e Vias Metabólicas/efeitos dos fármacos , Análise de Componente Principal , Selênio/química , Sulfóxidos , Regulação para Cima/efeitos dos fármacos
4.
J Pharmacol Exp Ther ; 343(2): 426-33, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22895897

RESUMO

Prostaglandin reductase 1 (PTGR1) is a highly inducible enzyme with enone reductase activity. Previous studies demonstrated the role of rat PTGR1 in the activation of acylfulvene analogs, a class of antitumor natural product derivatives. Of these, hydroxymethylacylfulvene (HMAF) was in advanced clinical development for the treatment of advanced solid tumors, including prostate, ovarian, and pancreatic cancers. However, the efficiency of human PTGR1 in activating acylfulvenes and its potential to enhance therapeutic efficacy have remained uncharacterized. In this study, human PTGR1 was polymerase chain reaction-cloned and purified. Conversion of HMAF to its cellular metabolite by the purified enzyme proceeded at a 20-fold higher rate than with the rat variant of the enzyme. The Km was 4.9 µM, which was 40-fold lower than for the rat variant and similar to the therapeutic dose. Human cell lines, including colon cancer lines, were transfected with a vector containing rat PTGR1 or human PTGR1, and cell viability was examined after dosing with HMAF. New data obtained in this study suggest that transfection with human PTGR1, or its induction in colon and liver cancer cell lines with 1,2-dithiol-3-thione, enhances susceptibility to the cytotoxic influences of HMAF by 2- to 10-fold. Furthermore, similar or enhanced enzyme induction and HMAF toxicity results from preconditioning cancer cells with the bioactive food components curcumin and resveratrol. The functional impact of PTGR1 induction in human cells and chemical-based strategies for its activation can provide important knowledge for the design of clinical strategies involving reductively activated cytotoxic chemotherapeutics.


Assuntos
15-Oxoprostaglandina 13-Redutase/biossíntese , Antineoplásicos Alquilantes/farmacologia , Sesquiterpenos/farmacologia , 15-Oxoprostaglandina 13-Redutase/genética , Animais , Elementos de Resposta Antioxidante , Antioxidantes/farmacologia , Biotransformação/fisiologia , Western Blotting , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Clonagem Molecular , Indução Enzimática/efeitos dos fármacos , Humanos , Indicadores e Reagentes , Cinética , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Plasmídeos/genética , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas/efeitos dos fármacos , Ratos , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Regulação para Cima/fisiologia
5.
Chimia (Aarau) ; 65(6): 411-5, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21797170

RESUMO

Chemical and biochemical processes underlying food-drug interactions in cancer therapy have not been well addressed with a systematic focus, even though they offer significant potential for enhancing the efficacy of cancer chemotherapy. Bioreductive anticancer drugs are metabolically activated by reductase enzymes. The levels and activities of relevant metabolic enzymes are regulated by transcription factors, which are under the control of chemical interactions with small molecules, including bioactive food components (BFCs) such as minerals, vitamins, and a variety of phytochemicals. One important and well-established process is the upregulation of enzymes involved in xenobiotic metabolism and redox regulation. Thus, BFCs might help to overcome resistances of some cancer cells towards anticancer agents or to increase efficacy by sensitizing cancer cells towards synergistic drugs. By understanding chemical and biochemical processes involved in food-drug interactions, not only can the risk of harmful food-drug interactions be diminished, but appropriate nutritional recommendations for cancer patients can be made and new functional foods with specific benefits in anticancer therapy may be developed.


Assuntos
Antineoplásicos/farmacologia , Interações Alimento-Droga , Neoplasias/tratamento farmacológico , Estado Nutricional , Antineoplásicos/metabolismo , Humanos , Oxirredutases/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA