Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Pflugers Arch ; 467(6): 1249-59, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25017107

RESUMO

Peroxisome proliferator-activated receptor ß (PPARß) and NaV1.5 voltage-gated sodium channels have independently been shown to regulate human breast cancer cell invasiveness. The n-3 polyunsaturated docosahexaenoic acid (DHA, 22:6n-3), a natural ligand of PPAR, is effective in increasing survival and chemotherapy efficacy in breast cancer patient with metastasis. DHA reduces breast cancer cell invasiveness and it also inhibits PPARß expression. We have shown previously that NaV1.5 promotes MDA-MB-231 breast cancer cells invasiveness by potentiating the activity of Na(+)/H(+) exchanger type 1 (NHE-1), the major regulator of H(+) efflux in these cells. We report here that DHA inhibited NaV1.5 current and NHE-1 activity in human breast cancer cells, and in turn reduced NaV1.5-dependent cancer cell invasiveness. For the first time, we show that antagonizing PPARß, or inhibiting its expression, reduced NaV1.5 mRNA and protein expression and NaV1.5 current, as well as NHE-1 activity and cell invasiveness. Consistent with these results, the DHA-induced reduction of both NaV1.5 expression and NHE-1 activity was abolished in cancer cells knocked-down for the expression of PPARß (shPPARß). This demonstrates a direct link between the inhibition of PPARß expression and the inhibition of Nav1.5/NHE-1 activities and breast cancer cell invasiveness. This study provides new mechanistic data advocating for the use of natural fatty acids such as DHA to block the development of breast cancer metastases.


Assuntos
Movimento Celular/efeitos dos fármacos , Ácidos Docosa-Hexaenoicos/farmacologia , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , PPAR beta/metabolismo , Linhagem Celular Tumoral , Humanos , Canal de Sódio Disparado por Voltagem NAV1.5/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
2.
J Cell Sci ; 126(Pt 21): 4835-42, 2013 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-23902689

RESUMO

The degradation of the extracellular matrix by cancer cells represents an essential step in metastatic progression and this is performed by cancer cell structures called invadopodia. NaV1.5 (also known as SCN5A) Na(+) channels are overexpressed in breast cancer tumours and are associated with metastatic occurrence. It has been previously shown that NaV1.5 activity enhances breast cancer cell invasiveness through perimembrane acidification and subsequent degradation of the extracellular matrix by cysteine cathepsins. Here, we show that NaV1.5 colocalises with Na(+)/H(+) exchanger type 1 (NHE-1) and caveolin-1 at the sites of matrix remodelling in invadopodia of MDA-MB-231 breast cancer cells. NHE-1, NaV1.5 and caveolin-1 co-immunoprecipitated, which indicates a close association between these proteins. We found that the expression of NaV1.5 was responsible for the allosteric modulation of NHE-1, rendering it more active at the intracellular pH range of 6.4-7; thus, it potentially extrudes more protons into the extracellular space. Furthermore, NaV1.5 expression increased Src kinase activity and the phosphorylation (Y421) of the actin-nucleation-promoting factor cortactin, modified F-actin polymerisation and promoted the acquisition of an invasive morphology in these cells. Taken together, our study suggests that NaV1.5 is a central regulator of invadopodia formation and activity in breast cancer cells.


Assuntos
Neoplasias da Mama/metabolismo , Extensões da Superfície Celular/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Neoplasias da Mama/genética , Caveolina 1/genética , Caveolina 1/metabolismo , Linhagem Celular Tumoral , Extensões da Superfície Celular/genética , Cortactina/genética , Cortactina/metabolismo , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Feminino , Humanos , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Fosforilação , Ligação Proteica , Trocadores de Sódio-Hidrogênio/genética
3.
Carcinogenesis ; 34(7): 1487-96, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23524196

RESUMO

The adenosine 5'-triphosphate (ATP)-gated Ca(2+)-permeable channel P2X7 receptor (P2X7R) is strongly upregulated in many tumors and cancer cells, and has an important role in cancer cell invasion associated with metastases. Emodin (1,3,8-trihydroxy-6-methylanthraquinone) is an anthraquinone derivative originally isolated from Rheum officinale Baill known for decades to possess anticancer properties. In this study, we examined the effects of emodin on P2X7R-dependent Ca(2+) signaling, extracellular matrix degradation, and in vitro and in vivo cancer cell invasiveness using highly aggressive human cancer cells. Inclusion of emodin at doses ≤10 µM in cell culture had no or very mild effect on the cell viability. ATP elicited increases in intracellular Ca(2+) concentration were reduced by 35 and 60% by 1 and 10 µM emodin, respectively. Emodin specifically inhibited P2X7R-mediated currents with an IC50 of 3 µM and did not inhibit the currents mediated by the other human P2X receptors heterologously expressed in human embryonic kidney (HEK293T) cells. ATP-induced increase in gelatinolytic activity, in cancer cell invasiveness in vitro and in cell morphology changes were prevented by 1 µM emodin. Furthermore, such ATP-evoked effects and inhibition by emodin were almost completely ablated in cancer cells transfected with P2X7R-specific small interfering RNA (siRNA) but not with scrambled siRNA. Finally, the in vivo invasiveness of the P2X7R-positive MDA-MB-435s breast cancer cells, assessed using a zebrafish model of micrometastases, was suppressed by 40 and 50% by 1 and 10 µM emodin. Taken together, these results provide consistent evidence to indicate that emodin inhibits human cancer cell invasiveness by specifically antagonizing the P2X7R.


Assuntos
Apoptose , Emodina/farmacologia , Invasividade Neoplásica/prevenção & controle , Antagonistas do Receptor Purinérgico P2X/farmacologia , Receptores Purinérgicos P2X7/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Antineoplásicos Fitogênicos/farmacologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Cálcio/metabolismo , Sinalização do Cálcio , Forma Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Feminino , Células HEK293 , Humanos , Concentração Inibidora 50 , Modelos Animais , Invasividade Neoplásica/patologia , Metástase Neoplásica/tratamento farmacológico , Metástase Neoplásica/patologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Rheum/química , Transfecção , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo
4.
Eur J Cell Biol ; 91(11-12): 847-60, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22673002

RESUMO

Invadosomes are actin-rich finger-like cellular structures sensing and interacting with the surrounding extracellular matrix (ECM) and involved in its proteolytic remodeling. Invadosomes are structures distinct from other adhesion complexes, and have been identified in normal cells that have to cross tissue barriers to fulfill their function such as leukocytes, osteoclasts and endothelial cells. They also represent features of highly aggressive cancer cells, allowing them to escape from the primary tumor, to invade surrounding tissues and to reach systemic circulation. They are localized to the ventral membrane of cells grown under 2-dimensional conditions and are supposed to be present all around cells grown in 3-dimensional matrices. Indeed invadosomes are key structures in physiological processes such as inflammation and the immune response, bone remodeling, tissue repair, but also in pathological conditions such as osteopetrosis and the development of metastases. Invadosomes are subdivided into podosomes, found in normal cells, and into invadopodia specific for cancer cells. While these two structures exhibit differences in organization, size, number and half-life, they share similarities in molecular composition, participation in cell-matrix adhesion and promoting matrix degradation. A key determinant in invadosomal function is the recruitment and release of proteases, such as matrix metalloproteinases (MMPs), serine proteases and cysteine cathepsins, together with their activation in a tightly controlled and highly acidic microenvironment. Therefore numerous pH regulators such as V-ATPases and Na(+)/H(+) exchangers, are found in invadosomes and are directly involved in their constitution as well as their functioning. This review focuses on the participation of pH regulators in invadosome function in physiological and pathological conditions, with a particular emphasis on ECM remodeling by osteoclasts during bone resorption and by cancer cells.


Assuntos
Estruturas da Membrana Celular/metabolismo , Matriz Extracelular/metabolismo , Prótons , Animais , Adesão Celular , Humanos , Concentração de Íons de Hidrogênio , Transporte de Íons , Osteoclastos/metabolismo , Peptídeo Hidrolases/metabolismo
5.
J Biol Chem ; 284(13): 8680-91, 2009 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-19176528

RESUMO

Voltage-gated sodium channels (Na(V)) are functionally expressed in highly metastatic cancer cells derived from nonexcitable epithelial tissues (breast, prostate, lung, and cervix). MDA-MB-231 breast cancer cells express functional sodium channel complexes, consisting of Na(V)1.5 and associated auxiliary beta-subunits, that are responsible for a sustained inward sodium current at the membrane potential. Although these channels do not regulate cellular multiplication or migration, their inhibition by the specific blocker tetrodotoxin impairs both the extracellular gelatinolytic activity (monitored with DQ-gelatin) and cell invasiveness leading to the attenuation of colony growth and cell spreading in three-dimensional Matrigel-composed matrices. MDA-MB-231 cells express functional cysteine cathepsins, which we found play a predominant role ( approximately 65%) in cancer invasiveness. Matrigel invasion is significantly decreased in the presence of specific inhibitors of cathepsins B and S (CA-074 and Z-FL-COCHO, respectively), and co-application of tetrodotoxin does not further reduce cell invasion. This suggests that cathepsins B and S are involved in invasiveness and that their proteolytic activity partly depends on Na(V) function. Inhibiting Na(V) has no consequence for cathepsins at the transcription, translation, and secretion levels. However, Na(V) activity leads to an intracellular alkalinization and a perimembrane acidification favorable for the extracellular activity of these acidic proteases. We propose that Na(v) enhance the invasiveness of cancer cells by favoring the pH-dependent activity of cysteine cathepsins. This general mechanism could lead to the identification of new targets allowing the therapeutic prevention of metastases.


Assuntos
Catepsina B/metabolismo , Catepsinas/metabolismo , Proteínas Musculares/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Canais de Sódio/metabolismo , Sódio/metabolismo , Catepsina B/antagonistas & inibidores , Catepsinas/antagonistas & inibidores , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Inibidores de Cisteína Proteinase/farmacologia , Dipeptídeos/farmacologia , Humanos , Concentração de Íons de Hidrogênio , Transporte de Íons/efeitos dos fármacos , Proteínas Musculares/antagonistas & inibidores , Canal de Sódio Disparado por Voltagem NAV1.5 , Invasividade Neoplásica , Proteínas de Neoplasias/antagonistas & inibidores , Neoplasias/patologia , Bloqueadores dos Canais de Sódio/farmacologia , Tetrodotoxina
6.
Free Radic Biol Med ; 44(7): 1483-91, 2008 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-18267129

RESUMO

Docosahexaenoic acid (DHA, a lipid of marine origin) has been found to enhance the activity of several anticancer drugs through an oxidative mechanism. To examine the relation between chemosensitization by DHA and tumor cells antioxidant status, we used two breast cancer cell lines: MDA-MB-231, in which DHA increases sensitivity to doxorubicin, and MCF-7, which does not respond to DHA. Under these conditions, reactive oxygen species (ROS) level increased on anthracycline treatment only in MDA-MB-231. This was concomitant with a decreased cytosolic glutathione peroxidase (GPx1) activity, a crucial enzyme for protection against hydrogen and lipid peroxides, while major antioxidant enzyme activities increased in both cell lines in response to ROS. GPx-decreased activity was accompanied by an accumulation of glutathione, the GPx cosubstrate, and resulted from a decreased amount of GPx protein. In rat mammary tumors, when a DHA dietary supplementation led to an increased tumor sensitivity to anthracyclines, GPx1 activity was similarly decreased. Furthermore, vitamin E abolished both DHA effects on chemotherapy efficacy enhancement and on GPx1 inhibition. Thus, loss of GPx response to an oxidative stress in transformed cells may account for the ability of peroxidizable targets such as DHA to enhance tumor sensitivity to ROS-generating anticancer drugs.


Assuntos
Antraciclinas/metabolismo , Neoplasias da Mama/metabolismo , Ácidos Docosa-Hexaenoicos/metabolismo , Regulação Neoplásica da Expressão Gênica , Glutationa Peroxidase/metabolismo , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Feminino , Glutationa/metabolismo , Humanos , Neoplasias Mamárias Animais/tratamento farmacológico , Estresse Oxidativo , Ratos , Espécies Reativas de Oxigênio
7.
Eur J Med Chem ; 43(5): 906-16, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-17692435

RESUMO

The pharmacomodulation of the N atom of alpha,beta-acetylenic aminothiolesters or the replacement of the thiolester moiety by more electrophilic groups did not permit any clear rationale to be established for improving the selective growth-inhibitory activity of this family of compounds over that of the previously synthesized alpha,beta-acetylenic aminothiolesters DIMATE and MATE [G. Quash, G. Fournet, J. Chantepie, J. Goré, C. Ardiet, D. Ardail, Y. Michal, U. Reichert, Biochem Pharmacol 64 (2002) 1279-92]. Hence DIMATE and MATE were investigated more thoroughly for selectivity and growth-inhibitory activity using human prostate epithelial normal cells (HPENC) on the one hand and human prostate epithelial cancer cells (DU145) on the other. Unequivocal evidence was obtained showing that both compounds were reversible growth inhibitors of HPENC but irreversible growth inhibitors of DU145. Growth-inhibition of DU145 was due to the induction of early apoptosis as revealed by the flow cytometric analytical profile of inhibitor-treated cells, of the decrease in the redox potential and increase in superoxide anion content of their mitochondria. Of the two intracellular enzymes: aldehyde dehydrogenases 1 and 3 (ALDH1 and ALDH3) targeted by DIMATE and MATE, ALDH3 was inhibited to the same extent by both compounds whereas ALDH1 was less susceptible to inhibition by MATE. As the induction of ALDH3 by xenobiotics is hormone-dependent, MATE, the more selective of the two inhibitors, is a useful tool not only for examining the role of the ALDH3 isoform in hormone-sensitive and resistant prostate cancer cells in culture but also for investigating if it can inhibit the growth of xenografts of prostate cancer in immunodeficient mice.


Assuntos
Aldeído Desidrogenase/antagonistas & inibidores , Antineoplásicos/síntese química , Apoptose , Células Epiteliais/efeitos dos fármacos , Compostos de Sulfidrila/síntese química , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Células Epiteliais/citologia , Ésteres , Humanos , Isoenzimas/antagonistas & inibidores , Masculino , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Oxirredução , Neoplasias da Próstata , Relação Estrutura-Atividade , Compostos de Sulfidrila/química , Compostos de Sulfidrila/farmacologia , Superóxidos/metabolismo , Transplante Heterólogo
8.
Drug Metab Dispos ; 35(5): 822-8, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17296624

RESUMO

The present work investigates the relationship between cancer cell chemosensitivity and subcellular distribution, molecular interaction, and metabolism of an anticancer drug. To get insights into this relationship, we took advantage of the differential sensitivity of two breast cancer cell lines, MDA-MB-231 and MCF-7, to anthracyclines, along with the property of docosahexaenoic acid (DHA, 22:6n-3), to differentially enhance their cytotoxic activity. The fluorescent drug mitoxantrone (MTX) was used because of the possibility to study its subcellular accumulation by confocal spectral imaging (CSI). The use of CSI allowed us to obtain semiquantitative maps of four intracellular species: nuclear MTX bound to DNA, MTX oxidative metabolite in endoplasmic reticulum, cytosolic MTX, and finally, MTX in a low polarity environment characteristic of membranes. MDA-MB-231 cells were found to be more sensitive to MTX (IC50 = 18 nM) than MCF-7 cells (IC50 = 196 nM). According to fluorescence levels, the nuclear and cytosolic MTX content was higher in MCF-7 than in MDA-MB-231 cells, indicating that mechanisms other than nuclear MTX accumulation account for chemosensitivity. In the cytosol, the relative proportion of oxidized MTX was higher in MDA-MB-231 (60%) than in MCF-7 (7%) cells. DHA sensitized MDA-MB-231 (approximately 4-fold) but not MCF-7 cells to MTX and increased MTX accumulation by 1.5-fold in MDA-MB-231 cells only. The DHA-stimulated accumulation of MTX was attributed mainly to the oxidative metabolite. Antioxidant N-acetyl-L-cysteine inhibited the DHA effect on both metabolite accumulation and cell sensitization to MTX. We conclude that drug metabolism and compartmentalization are associated with cell chemosensitization, and the related cytotoxicity mechanisms may involve oxidative stress.


Assuntos
Núcleo Celular/metabolismo , Citosol/metabolismo , Mitoxantrona/farmacologia , Mitoxantrona/farmacocinética , Acetilcisteína/farmacologia , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Ácidos Docosa-Hexaenoicos/farmacologia , Feminino , Corantes Fluorescentes/metabolismo , Humanos , Concentração Inibidora 50 , Microscopia Confocal , Mitoxantrona/química , Estrutura Molecular , Espécies Reativas de Oxigênio/metabolismo , Rodamina 123/metabolismo , Fatores de Tempo
9.
Bioorg Chem ; 34(1): 49-58, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16387348

RESUMO

6S,8S-Bis(3-methylthiopropanoyl) thiolesters of lipoic acid were synthesized with the carboxyl moiety of lipoate modified as methyl or water soluble choline esters. Evaluation on different cell lines in culture showed that they possessed modest antiproliferative activity. However, the 6-fold decrease in IC50 (from 270 to 45 microM) observed with the water soluble 6S,8S-bis(3-methylthiopropenoyl) thiolester dehydro derivative on a human epithelial prostate cancer cell line (DU145) argues in favor of 3-methylthiopropanoyl metabolites as endogenous growth regulatory (apoptogenic) compounds derived from methionine.


Assuntos
Aldeídos/química , Antineoplásicos/síntese química , Ésteres/síntese química , Propionatos/química , Ácido Tióctico/síntese química , Aldeídos/metabolismo , Antineoplásicos/farmacologia , Ésteres/farmacologia , Humanos , Concentração Inibidora 50 , Masculino , Metionina/química , Mimetismo Molecular , Neoplasias da Próstata/patologia , Compostos de Sulfidrila/química , Ácido Tióctico/farmacologia , Células Tumorais Cultivadas
10.
Free Radic Biol Med ; 39(6): 742-51, 2005 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-16109304

RESUMO

Polyunsaturated fatty acids have been reported to enhance the cytotoxic activity of several anticancer drugs. In the present study, we observed that doxorubicin chemosensitization of breast cancer cell lines by docosahexaenoic acid (DHA, a long-chain omega-3 polyunsaturated fatty acid) was cell-line selective, affecting MDA-MB-231 and MCF-7 dox (a doxorubicin-resistant cell line) but not the parental MCF-7 cell line. DHA supplementation led to an increase in membrane phospholipid DHA level, but did not induce changes in intracellular [(14)C]doxorubicin accumulation. In MDA-MB-231, doxorubicin efficacy enhancement by DHA was linked to an increase in malondialdehyde level, a final product of lipid peroxidation. DHA elicited by itself a 3.7-fold malondialdehyde level increase, additive to that induced by doxorubicin. Addition of doxorubicin to DHA further increased the glutathione level, indicative of the generation of an oxidative stress. In contrast to MDA-MB-231, doxorubicin did not increase the malondialdehyde level in MCF-7, although DHA induced lipid peroxidation. Therefore in MCF-7, lipid peroxidation induced by DHA itself was not sufficient to trigger an oxidative stress and to subsequently increase sensitivity to doxorubicin. These data indicate that the differential effect of DHA among cells on drug toxicity results from a differential oxidative response to doxorubicin. Chemosensitization through fatty acids appears as a new promising adjuvant therapeutic paradigm, since omega-3 fatty acids are physiological molecules found in food and are nontoxic in vivo.


Assuntos
Ácidos Docosa-Hexaenoicos/farmacologia , Doxorrubicina/farmacologia , Sinergismo Farmacológico , Neoplasias/tratamento farmacológico , Antibióticos Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Quimioterapia Adjuvante , Ácidos Docosa-Hexaenoicos/química , Doxorrubicina/química , Ácidos Graxos/metabolismo , Ácidos Graxos Ômega-3/metabolismo , Glutationa/química , Glutationa/metabolismo , Dissulfeto de Glutationa/metabolismo , Humanos , Peroxidação de Lipídeos , Malondialdeído/metabolismo , Oxirredução , Oxigênio/metabolismo , Fosfolipídeos/química , Fatores de Tempo , Vitamina E/metabolismo
11.
Biochem Pharmacol ; 64(8): 1279-92, 2002 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-12234608

RESUMO

4-Amino-4-methyl-pent-2-ynthioc acid S-methyl ester (ampal thiolester: ATE) was used as a lead compound to synthesise new amino-substituted derivatives of alpha, beta acetylenic thiolester compounds as inhibitors of aldehyde dehydrogenase 1, (ALDH1). Of these compounds, the dimethyl derivative (DIMATE) was a competitive irreversible inhibitor (K(i) approximately 280 microM) of baker's yeast ALDH1 in vitro showing 80% inhibition at 400 microM when preincubated with the enzyme for 30min, whereas the trimethyl ammonium and the morpholine derivatives showed only 15% inhibition at 600 microM even after 60min preincubation. ATE inhibited ALDH1 activity in ALDH1-transfected L1210 T cells resistant to hydroperoxycyclophosphamide (HCPA) and inhibited growth synergistically in the presence of HCPA. In non-transfected L1210 counterparts ATE did not potentiate growth inhibition by HCPA. DIMATE was a 30-100-fold more effective growth inhibitor than ATE. Endogenous ALDH1 activities of BAF(3) cells over-expressing different levels of bcl(2) (0-100%) were similar (16-20mU/mg protein) and were all inhibited by DIMATE, reaching 20-30% at 4 microM. Up to 4 microM no apoptosis, as measured by DNA-fragmentation was observed, but at 8 and 10 microM DIMATE, DNA-fragmentation increased concomitantly with ALDH1 inhibition. No DNA-fragmentation was observed with ALDH1 irreversible inhibitors devoid of a thiolester group or with thiolesters which were not inhibitors of ALDH1. It was seen only with competitive irreversible inhibitors having the methanethiol and enzyme-inhibitory moieties. The methanethiol putatively released from DIMATE by ALDH1 esterase activity plays a role, albeit undefined, in lowering intramitochondrial glutathione levels which decreased by 47% as DNA-fragmentation increased.


Assuntos
Aldeído Desidrogenase/antagonistas & inibidores , Apoptose , Ciclofosfamida/análogos & derivados , Isoenzimas/antagonistas & inibidores , Leucemia L1210/patologia , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Aldeído Desidrogenase/metabolismo , Família Aldeído Desidrogenase 1 , Alcinos/farmacologia , Animais , Ciclofosfamida/farmacologia , Fragmentação do DNA/efeitos dos fármacos , Ésteres/farmacologia , Glutationa/metabolismo , Humanos , Isoenzimas/metabolismo , Linfócitos/efeitos dos fármacos , Linfócitos/metabolismo , Camundongos , Proteínas Proto-Oncogênicas c-bcl-2/fisiologia , Retinal Desidrogenase , Saccharomyces cerevisiae/enzimologia , Compostos de Sulfidrila/farmacologia , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA