Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Am J Physiol Cell Physiol ; 281(5): C1579-86, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11600421

RESUMO

Intestinal absorption of folates has been characterized as a facilitative process with a low pH optimum. Studies with intestinal epithelial cells have suggested that this activity is mediated by the reduced folate carrier (RFC1). In this paper, we report on folate transport characteristics in an immortalized rat IEC-6 cell line that was found to exhibit the predominant influx activity for methotrexate (MTX) at pH 5.5 with a low level of activity at pH 7.4. Transfection of this cell line with an RFC1 construct resulted in clones exhibiting increased MTX uptake at both the pHs and high folic acid uptake only at the low pH. For the two clones with the highest level of transport activity, relative MTX influx at the two pHs was reversed. Moreover, the low pH MTX influx activity ([MTX](e) = 0.5 microM) was markedly inhibited by 20 microM folic acid while influx at neutral pH was not. Furthermore, in the presence and absence of glucose at low pH, MTX and folic acid influx activity was inhibited by azide, while MTX influx at pH 7.4 was stimulated by azide in the absence of glucose but was unchanged in the presence of glucose and azide. This was contrasted with the results of transfection of the same RFC1 construct into an L1210 murine leukemia cell line bearing a nonfunctional endogenous carrier. In this case, the activity expressed was only at pH 7.4. These data indicate that RFC1 can exhibit two distinct types of folate transport activities in intestinal cells that must depend on tissue-specific modulators.


Assuntos
Ácido Fólico/metabolismo , Proteínas de Membrana Transportadoras/biossíntese , Animais , Transporte Biológico Ativo , Northern Blotting , Western Blotting , Linhagem Celular , Clonagem Molecular , Metabolismo Energético/efeitos dos fármacos , Células Epiteliais/metabolismo , Ácido Fólico/farmacologia , Antagonistas do Ácido Fólico/metabolismo , Antagonistas do Ácido Fólico/farmacologia , Concentração de Íons de Hidrogênio , Proteínas de Membrana Transportadoras/genética , Metotrexato/metabolismo , Metotrexato/farmacologia , Oxirredução , Ratos , Transfecção
2.
Semin Oncol ; 26(2 Suppl 6): 11-23, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10598550

RESUMO

Transport of folates and antifolates in most cells is mediated by the reduced folate carrier (RFCI), an anion exchanging concentrative process, which is opposed by independent exit pump(s) that are directly coupled to energy metabolism. The balance of these processes governs the free intracellular folate level that is the substrate for folylpolyglutamate synthetase and determines the rate of metabolism of many folates and antifolates to polyglutamate derivatives. The RFCI has a high affinity for new-generation antifolates, but the relationship between transport and polyglutamylation for all these compounds has not, as yet, been defined. Analysis of mutations in RFCI indicate highly selective functional changes in binding affinity and/or carrier mobility, raising the possibilities that (1) enhanced transport and accumulation of natural folates can result in resistance to antifolate inhibitors of purine and pyrimidine synthesis by blocking their polyglutamylation to active derivatives, or by competition at the target enzyme site without substantial changes in antifolate transport, and (2) transport-related resistance to one antifolate might not produce comparable cross-resistance to another. Impaired export pump function, which results in enhanced accumulation of folates in cells and inhibition of antifolate polyglutamylation, represents another potential mechanism of resistance to new-generation antifolates. Folate receptor-mediated transport is another route of entry of folates and antifolates into cells, but its importance has not been well-defined. Finally, a low pH transport mechanism is present in a variety of mammalian cell lines. However, its relationship to RFCI and other low pH epithelial transporters and its ability to transport new-generation antifolates remain to be established.


Assuntos
Transporte Biológico/fisiologia , Proteínas de Transporte/fisiologia , Antagonistas do Ácido Fólico/metabolismo , Ácido Fólico/metabolismo , Proteínas de Membrana , Proteínas de Membrana Transportadoras , Animais , Antimetabólitos Antineoplásicos/farmacologia , Proteínas de Transporte/genética , Membrana Celular/fisiologia , Resistencia a Medicamentos Antineoplásicos , Antagonistas do Ácido Fólico/farmacologia , Humanos , Concentração de Íons de Hidrogênio , Metotrexato/farmacologia , Força Próton-Motriz , Proteína Carregadora de Folato Reduzido , Relação Estrutura-Atividade
3.
Biochem Pharmacol ; 58(8): 1321-7, 1999 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-10487535

RESUMO

Studies are reported that describe the multifaceted inhibitory effects of prostaglandin A1 (PGA1) on processes that govern the transport of folates across the plasma membrane of Chinese hamster ovary (CHO) cells: the reduced folate carrier, RFC1, and ATP-dependent exporters. PGA1 was a noncompetitive inhibitor of MTX influx mediated by RFC1 with a Ki of approximately 21 microM. The onset of inhibition was virtually instantaneous, not reversible, and appeared to require the incorporation of PGA1 into the lipid membrane; surface adsorption alone was insufficient for inhibition of RFC1 transport activity. In contrast, the effect of PGA1 on folic acid transport was small (approximately 20% inhibition of total influx), consistent with the observation that the major portion of folic acid transport in CHO cells is mediated by a low pH mechanism distinct from RFC1. PGA1 was also a potent inhibitor of the ATP-driven efflux of both MTX and folic acid. At a concentration of 7 microM PGA1, the efflux rate constants for these folates were depressed by approximately 70 and approximately 50%, respectively. The net effects of PGA1 on the bidirectional folate fluxes translated into marked alterations in net transport. The addition of 7 microM PGA1 to cells at steady state with 1 microM MTX produced a rapid onset of net uptake and the achievement of an approximately 3-fold increase in the steady-state free MTX level as compared with untreated CHO cells. The addition of 7 microM PGA1 to cells at steady state with 1 microM folic acid produced an approximately 5-fold increase in the free folate level. These studies establish PGA1 as a potent inhibitor of both the reduced folate carrier and ATP-driven folate exporter(s). The noncompetitive nature of the inhibition of RFC1 is unique among anionic compounds, which are usually competitive inhibitors of the carrier.


Assuntos
Proteínas de Transporte/antagonistas & inibidores , Membrana Celular/efeitos dos fármacos , Ácido Fólico/metabolismo , Prostaglandinas A/farmacologia , Receptores de Superfície Celular , Trifosfato de Adenosina/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Células CHO , Membrana Celular/metabolismo , Cricetinae , Receptores de Folato com Âncoras de GPI , Antagonistas do Ácido Fólico/farmacocinética , Metotrexato/farmacocinética
4.
Biochem Pharmacol ; 55(9): 1505-12, 1998 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-10076544

RESUMO

Folate influx at low pH was characterized in MTXrA cells, an L1210 mouse leukemia cell line with a functional defect in the reduced folate carrier. Folic acid influx in MTXrA cells was negligible at pH 7.5, increased 13-fold as the pH was decreased to 6.0, and was indistinguishable from that in L1210 cells. In contrast, while methotrexate (MTX) influx in MTXrA cells at pH 6.0 was 15-fold higher than at pH 7.5, in L1210 cells it was decreased by half. Influx of MTX, folic acid, 5-methyltetrahydrofolate and 5-formyltetrahydrofolate in MTXrA cells was increased at pH < 7.0, but their pH optima and profile differed substantially. Influx of MTX and 5-methyltetrahydrofolate at pH 6.0 showed saturability, with a Kt of 2.65 and 0.56 microM, and a Vmax of 0.45 and 0.083 nmol/g dry wt/min, respectively. MTX influx mediated by the low pH transporter was insensitive to the anionic composition of the transport buffer and affected minimally (approximately 20%) by Na+ substitution. The anion transport inhibitors sulfobromophthalein, diisothiocyanatostilbene disulfonic acid, and acetamidoisothiocyanatostilbene disulfonic acid were not effective inhibitors of the low pH route. MTX transport at low pH did not increase in MTXrA-R16 cells, an MTXrA derivative with 10-fold overexpression of the reduced folate carrier (RFC) due to transfection with RFC1 cDNA. Inhibition of reduced folate carrier activity with acetamidoisothiocyanatostilbene disulfonic acid resulted in identical MTX influx in L1210, MTXrA, and MTXrA-R16 cells at pH 5.5. Finally, low pH-mediated MTX influx was reduced by energy inhibitors and partially inhibited by ionophores (nigericin > monensin >> valinomycin). The data indicate that L1210 and MTXrA cells express similar activities of a low pH folate transporter that has properties distinct from, and independent of, the reduced folate carrier.


Assuntos
Proteínas de Transporte/metabolismo , Antagonistas do Ácido Fólico/farmacocinética , Ácido Fólico/metabolismo , Concentração de Íons de Hidrogênio , Leucemia L1210/metabolismo , Metotrexato/farmacocinética , Receptores de Superfície Celular , Ácido 4-Acetamido-4'-isotiocianatostilbeno-2,2'-dissulfônico/farmacologia , Cloreto de Amônio/farmacologia , Animais , Transporte Biológico/efeitos dos fármacos , Carbonil Cianeto m-Clorofenil Hidrazona/farmacologia , Proteínas de Transporte/genética , Receptores de Folato com Âncoras de GPI , Cinética , Camundongos , Monensin/farmacologia , Nigericina/farmacologia , Células Tumorais Cultivadas , Valinomicina/farmacologia
5.
Biochim Biophys Acta ; 1353(2): 191-8, 1997 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-9294013

RESUMO

RT-PCR analysis of the reduced folate carrier (RFC) from L1210 and murine erythroleukemia cells led to the identification of three clones which appeared to result from the use of alternative splice sites. The nucleotide sequence of each splice form predicts a protein that contains at least the first 7 transmembrane domains of the parental RFC protein followed by a novel hydrophilic carboxyl terminus of 33, 72, or 105 amino acid residues. Sequence analysis of cDNA clones isolated from murine liver and the results of 5'-RACE from L1210 cells indicated that RFC also utilizes alternate 5'-terminal exons. To understand how the alternatively spliced RFC transcripts and multiple 5'-termini were generated, the genomic organization of RFC was determined. The gene is comprised of at least 8 exons, the first two of which encode the alternative 5' termini. Based on sequence identity with cDNAs encoding RFC from hamster and rat, however, it appears that additional 5' exons may be present. Two of the RFC splice variants result from the use of a cryptic splice donor site within exon 4 and the third results from the use of a cryptic splice acceptor site within exon 5. In addition, the splice variant form that encodes the largest protein also utilizes an alternative exon located between exons 5 and 6. The apparent use of alternative transcriptional start sites and the identification of several RFC splice forms raises the possibility that unique RFC molecules may be generated that exhibit tissue- or cell line-specific distribution.


Assuntos
Processamento Alternativo , Proteínas de Transporte/genética , Proteínas de Membrana , Proteínas de Membrana Transportadoras , Animais , Sequência de Bases , Proteínas de Transporte/química , Clonagem Molecular , DNA Complementar/química , DNA Complementar/isolamento & purificação , Éxons , Biblioteca Genômica , Fígado/metabolismo , Camundongos , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Proteína Carregadora de Folato Reduzido , Células Tumorais Cultivadas
6.
Biochem Pharmacol ; 53(2): 223-31, 1997 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-9037255

RESUMO

F2-MTX'A is an L1210 leukemia cell line with a functional detect in the reduced folate carrier and high level expression of folate receptor beta. The pH dependence of methotrexate (MTX) influx by folate receptor beta in F2-MTX'A cells was characterized and compared with that of the reduced folate carrier in parental L1210 cells. MTX influx by folate receptor beta had a pH optimum of 6.5, whereas influx mediated by the reduced folate carrier showed a pH optimum of 7.5. Increased folate receptor beta-mediated MTX influx at pH 6.5 relative to pH 7.5 was accompanied by a 5-fold increase in binding affinity of the receptor for MTX without a change in the number of binding sites. At pH 6.2, approximately 24% of MTX influx in F2-MTX'A cells proceeded by another mechanism. This transport route became active at pH < 7.5, operated optimally at pH 6.0 to 6.5, and, unlike folate receptor beta-mediated MTX influx, was insensitive to the presence of low levels of folic acid (100 nM). MTX influx by the low pH system showed saturability, with a Ki of 5.3 microM and a Vmax of 1.53 nmol/g dry wt/min, was energy dependent, was inhibited by sulfobromophthalein with a Ki of 148 microM, and had similar relative affinities for folic acid, leucovorin, and 5 methyltetrahydrofolate. Influx of 5-methyltetrahydrofolate was also mediated by this route. The data provide further confirmatory evidence for an MTX influx route in F2-MTX'A cells, optimal at low pH and distinct from the reduced folate carrier or the folate receptor.


Assuntos
Proteínas de Transporte/fisiologia , Antagonistas do Ácido Fólico/farmacocinética , Leucemia L1210/metabolismo , Proteínas de Membrana , Proteínas de Membrana Transportadoras , Metotrexato/farmacocinética , Receptores de Superfície Celular , Animais , Transporte Biológico , Divisão Celular/efeitos dos fármacos , Receptores de Folato com Âncoras de GPI , Concentração de Íons de Hidrogênio , Metotrexato/farmacologia , Camundongos , Proteína Carregadora de Folato Reduzido , Sulfobromoftaleína/farmacologia , Células Tumorais Cultivadas
7.
Biochem Pharmacol ; 52(5): 703-12, 1996 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-8765468

RESUMO

We previously described a methotrexate-resistant L1210 cell line (MTXrA) that lacks a functional reduced folate carrier and does not appreciably express the folate receptor. In the present study, we utilized MTXrA cell lines stably transfected with cDNAs encoding either the folate receptor or the reduced folate carrier to investigate the influence of the route of folate influx on the rate and extent of methotrexate polyglutamylation. At an extracellular methotrexate concentration of 0.1 microM, influx in the folate receptor transfectant (MTXrA-TF1) and in the reduced folate carrier transfectant (MTXrA-R1) was equal and methotrexate polyglutamates accumulated at an identical rate, but the onset was delayed until dihydrofolate reductase was saturated with the monoglutamate (approxmately 3 hr). The onset of polyglutamate formation was immediate and identical among the lines in cells pretreated with the lipophilic dihydrofolate reductase inhibitor trimetrexate to block methotrexate binding to dihydrofolate reductase. The spectra of individual methotrexate polyglutamates that accumulated were similar, with the tetraglutamate present as the predominant form. A 100-fold higher methotrexate concentration was required to detect methotrexate uptake and polyglutamylation in the transport defective parent MTXrA line, demonstrating that diffusion or an unidentified low affinity route also supports polyglutamylation. Since the folate receptor and the reduced folate carrier achieve nearly identical rates of polyglutamylation despite very different mechanisms of methotrexate delivery, the data suggest that transport-mediated substrate channeling to folylpolyglutamate synthetase is unlikely to play a role in tetrahydrofolate metabolism. This study supports the notion that it is the intracellular concentration of methotrexate achieved within the cell that drives polyglutamylation irrespective of its route of entry.


Assuntos
Proteínas de Transporte/fisiologia , Leucemia L1210/metabolismo , Proteínas de Membrana , Proteínas de Membrana Transportadoras , Metotrexato/análogos & derivados , Ácido Poliglutâmico/análogos & derivados , Receptores de Superfície Celular , Animais , Transporte Biológico , Receptores de Folato com Âncoras de GPI , Leucemia L1210/enzimologia , Leucemia L1210/patologia , Metotrexato/metabolismo , Camundongos , Peptídeo Sintases/metabolismo , Ácido Poliglutâmico/metabolismo , Proteína Carregadora de Folato Reduzido , Tetra-Hidrofolato Desidrogenase/metabolismo , Células Tumorais Cultivadas
8.
Biochem Pharmacol ; 50(8): 1287-94, 1995 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-7488246

RESUMO

This laboratory previously described an L1210 murine leukemia cell line with a functional defect in the reduced folate carrier and increased expression of folate receptor-beta (F2-MTXrA). This cell line was used to characterize methotrexate (MTX) influx mediated by folate receptor-beta and to compare this with influx mediated by the reduced folate carrier in L1210 parental cells. Influx of 0.2 microM MTX in F2-MTXrA cells was one-third that of L1210 cells and was abolished by very low concentrations of folic acid. Kinetic analysis revealed that MTX transport mediated by folate receptor-beta exhibited an influx kappa t one-third, and an influx Vmax one-fourth, that of the reduced folate carrier. Metabolic inhibitors markedly suppressed influx in F2-MTXrA cells but had no effect on MTX influx in L1210 cells. MTX influx in both cell lines was inhibited by the organic anions probenecid, sulfobromophthalein, and CI-920, but to a lesser extent in F2-MTXrA cells. The inhibitory effects of these anions on transport in F2-MTXrA cells could be attributed to their inhibition of MTX binding to the folate receptor. Although MTX influx in both cell lines was not sodium dependent, removal of extracellular chloride increased influx 2-fold in L1210 cells while markedly inhibiting influx in F2-MTXrA cells. Substitution of Cl- with isethionate or NO3- partially restored influx in the latter cells, whereas SO4(2-) was inhibitory. Anions enhanced MTX binding to folate receptor-beta with isethionate > SO4(2-) > Cl-. Decreasing the buffer pH to 6.2 produced a 69% reduction, and a 260% increase, in MTX influx in L1210 cells and F2-MTXrA cells, respectively. The data indicate that folate receptor-beta-mediated MTX influx has properties fundamentally different from transport mediated by the reduced folate carrier in terms of energy, ion, and pH dependence. There was no evidence indicating that these processes are functionally linked.


Assuntos
Proteínas de Transporte/metabolismo , Leucemia L1210/metabolismo , Proteínas de Membrana , Proteínas de Membrana Transportadoras , Receptores de Superfície Celular , Animais , Transporte Biológico/efeitos dos fármacos , Receptores de Folato com Âncoras de GPI , Concentração de Íons de Hidrogênio , Cinética , Metotrexato/metabolismo , Camundongos , Probenecid/farmacologia , Proteína Carregadora de Folato Reduzido , Sulfobromoftaleína/farmacologia , Células Tumorais Cultivadas
9.
J Biol Chem ; 270(39): 22974-9, 1995 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-7559435

RESUMO

This laboratory previously described an L1210 leukemia cell line (MTXrA) selected for resistance to methotrexate by virtue of impaired transport due to a functional defect in the translocation process. We now report on the sequence analysis of cDNAs encoding the reduced folate carrier from this line and identify a single mutation that results in the substitution of a proline for an alanine in a highly conserved transmembrane region of the protein. Transfection of the parental reduced folate carrier into MTXrA cells resulted in a cell line which exhibited a complete restoration of methotrexate uptake and an enhanced sensitivity to methotrexate. Northern analysis and specific [3H]MTX cell surface binding indicated that expression of the reduced folate carrier was elevated approximately 5-fold in the transfectant compared to parental and MTXrA cells. The MTX influx properties of the transfectant cell line were identical to those of the well characterized reduced folate carrier from parental L1210 cells in terms of: 1) patterns of sensitivity to competing folates, 2) sensitivity to the organic anion sulfobromophthalein, 3) lack of energy dependence, and 4) capacity for trans-stimulation. We also provide new data which suggests that the nucleotide sequence 5' of the predicted ATG initiation codon may encode additional protein information in the form of a leader sequence. Finally, we demonstrate that the MTXrA line has both the mutant and the parental reduced folate carrier alleles but that expression appears to be restricted to the mutant allele. Thus, the methotrexate transport phenotype and resultant drug resistance in this cell line result from genetic/regulatory events at both alleles.


Assuntos
Antimetabólitos Antineoplásicos/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Leucemia L1210/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras , Metotrexato/metabolismo , Mutação , Alelos , Sequência de Aminoácidos , Animais , Sequência de Bases , Células CHO , Proteínas de Transporte/química , Clonagem Molecular , Códon , Sequência Conservada , Cricetinae , Primers do DNA , Proteínas de Membrana/química , Camundongos , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Receptores de Superfície Celular/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Mapeamento por Restrição , Homologia de Sequência de Aminoácidos , Transfecção , Células Tumorais Cultivadas
10.
J Biol Chem ; 270(14): 7842-9, 1995 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-7713875

RESUMO

L1210 leukemia cells transport reduced folates and methotrexate via a well defined reduced folate carrier system and, in the absence of low folate selective pressure, do not express an alternate endocytotic route mediated by cell surface folate receptors. This laboratory previously described an L1210 leukemia cell line, MTXrA, with acquired resistance to methotrexate (MTX) due to the loss of mobility of the reduced folate carrier. We now report on the transfection of MTXrA with a cDNA encoding the murine homolog of the human folate receptor isoform of KB cells to produce MTXrA-TF1, which constitutively expresses high levels of FR-alpha. MTXrA-TF1 and L1210 cells were utilized to compare transport of methotrexate mediated by FR-alpha and the reduced folate carrier, respectively. Methotrexate influx in the two lines was similar when the extracellular level was 0.1 microM, but as the methotrexate concentration increased, influx via the reduced folate carrier increased in comparison to influx mediated by FR-alpha. Transport kinetics indicated both a approximately 20-fold lower influx Kb and Vmax for MTXrA-TF1 as compared to L1210 cells. The two cell lines exhibited distinct influx properties. Methotrexate influx in MTXrA-TF1 was markedly inhibited by 50 nM folic acid and metabolic poisons. In L1210 cells, 1.0 microM folic acid did not affect MTX influx, and metabolic poisons either had no effect on or increased methotrexate influx. Removal of extracellular chloride markedly inhibited transport in MTXrA-TF1 but stimulated influx in L1210 cells. When the pH was decreased to 6.2, methotrexate influx was not altered in MTXrA-TF1 but was reduced in L1210 cells. Probenecid and sulfobromophthalein inhibit methotrexate influx in both L1210 and MTXrA-TF1 cell lines; however, inhibition in MTXrA-TF1 could be accounted for on the basis of inhibition of methotrexate binding to FR-alpha. The data indicate that the reduced folate carrier and FR-alpha function independently and exhibit distinct properties. FR-alpha expressed at sufficient levels can mediate influx of MTX and folates into cells at rates comparable to the reduced folate carrier and hence has pharmacologic and physiologic importance.


Assuntos
Proteínas de Transporte/fisiologia , Ácido Fólico/metabolismo , Proteínas de Membrana , Proteínas de Membrana Transportadoras , Receptores de Superfície Celular/fisiologia , Alcenos/farmacologia , Animais , Transporte Biológico , Metabolismo Energético , Receptores de Folato com Âncoras de GPI , Concentração de Íons de Hidrogênio , Íons , Leucemia L1210 , Metotrexato/metabolismo , Metotrexato/farmacocinética , Camundongos , Polienos , Probenecid/farmacologia , Pironas , Proteína Carregadora de Folato Reduzido , Sulfobromoftaleína/farmacologia , Temperatura , Transfecção , Células Tumorais Cultivadas
11.
Chem Res Toxicol ; 7(5): 609-13, 1994.
Artigo em Inglês | MEDLINE | ID: mdl-7841338

RESUMO

The metabolism of the o-quinone derivative of estrone, 3,4-estrone quinone (3,4-EQ), has been investigated in human breast cancer cells. Unlike the p-quinone, diethylstilbestrol 4',4"-quinone, 3,4-EQ was not a substrate for the two-electron reduction catalyzed by the putative detoxifying enzyme, NAD(P)H:quinone reductase (DT diaphorase; DT D). Accordingly, the DNA damage induced by 3,4-EQ in human MCF-7 cells was not affected by an inhibitor of DT D. Although 3,4-EQ was not an apparent substrate for the two-electron reduction catalyzed by DT D, this o-quinone was a substrate for the one-electron reduction catalyzed by cytochrome P450 reductase. The one-electron reduction of 3,4-EQ catalyzed by cytochrome P450 reductase occurred in the face of a significant and potentially physiologically relevant spontaneous reduction of 3,4-EQ by NADPH. The impact of purified superoxide dismutase (SOD) upon the production of hydrogen peroxide produced as a consequence of 3,4-EQ metabolism was evaluated; surprisingly, SOD inhibited the hydrogen peroxide produced by this o-quinone. Possible reasons for the SOD-mediated inhibition of redox cycling of 3,4-EQ are discussed. In summary, important differences in the metabolism of 3,4-EQ vis-a-vis o- and p- quinones have been observed, and the implications of these differences are discussed.


Assuntos
Estrenos/metabolismo , Animais , Dano ao DNA , Dietilestilbestrol/metabolismo , Humanos , Peróxido de Hidrogênio/metabolismo , Técnicas In Vitro , Fígado/enzimologia , Fígado/metabolismo , NAD(P)H Desidrogenase (Quinona)/metabolismo , NADP/metabolismo , NADPH-Ferri-Hemoproteína Redutase/metabolismo , Oxirredução , Ratos , Frações Subcelulares/enzimologia , Frações Subcelulares/metabolismo , Superóxido Dismutase/metabolismo , Células Tumorais Cultivadas
12.
J Lab Clin Med ; 123(4): 506-14, 1994 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-8144999

RESUMO

The induction of heme oxygenase in cells under conditions of oxidative stress has been hypothesized to represent a cellular antioxidant defense mechanism. The objectives of this study were to characterize the induction of heme oxygenase by the oxidant stress-inducing quinone agent menadione (2-methyl-1,4-naphthoquinone) and to elucidate the roles of basal and induced heme oxygenase enzyme activities in menadione-induced DNA damage and growth inhibition in human MCF-7 cells. Time- and dose-dependent inductions of heme oxygenase messenger RNA and enzyme activity in menadione-treated MCF-7 cells were demonstrated. Intracellular and extracellular bilirubin concentrations were less than 100 nmol/L and were not altered when heme oxygenase was induced. The roles of the basal and induced heme oxygenase enzyme activities in menadione-mediated DNA damage were evaluated by means of the heme oxygenase competitive inhibitor tin protoporphyrin. Inhibition of the basal heme oxygenase enzyme activity by tin protoporphyrin resulted in a decrease in the number of menadione-induced DNA breaks and an attenuation of the cellular growth inhibition caused by menadione. Induced heme oxygenase did not protect MCF-7 cells from menadione-induced DNA breaks. Basal heme oxygenase enzyme activities in two cloned menadione-resistant cell lines were significantly less than that measured in a menadione-sensitive parental MCF-7 cell line. Collectively, these data do not support a protective role for basal or induced heme oxygenase enzyme activities against oxidant stress-related DNA strand breakage or cytotoxic effects engendered by menadione in human cells.


Assuntos
Heme Oxigenase (Desciclizante)/biossíntese , Heme Oxigenase (Desciclizante)/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Vitamina K/farmacologia , Bilirrubina/biossíntese , Dano ao DNA , Indução Enzimática/efeitos dos fármacos , Heme Oxigenase (Desciclizante)/genética , Humanos , Oxirredução/efeitos dos fármacos , RNA Mensageiro/efeitos dos fármacos , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/metabolismo
13.
Chem Res Toxicol ; 7(1): 23-8, 1994.
Artigo em Inglês | MEDLINE | ID: mdl-8155821

RESUMO

The o-quinone forms of 2,3- and 3,4-catechol estrogens have been implicated in the carcinogenicity of these hormones. The concomitant production of reactive oxygen species during reduction of the o-quinone estrogens has been inferred to play a mechanistic role in their mutagenic potential. Conclusive evidence documenting the production of hydrogen peroxide, the hydroxyl radical, and the estrone 3,4-semiquinone in estrone 3,4-quinone (3,4-EQ)-treated human breast cancer subcellular fractions was demonstrated in the absence of exogenously added catalysts. Subcellular fractions of MCF-7 cells treated with 3,4-EQ and NADPH, including nuclei, mitochondria, and microsomes, were shown to support significant amounts of hydrogen peroxide production. Hydrogen peroxide production in 3,4-EQ-treated cellular fractions and the chromosomal DNA damage induced in 3,4-EQ-treated MCF-7 cells were abolished by the addition of catalase. A significant and potentially physiologically relevant spontaneous reduction of 3,4-EQ by NADPH resulting in hydrogen peroxide production was demonstrated. The results unequivocally demonstrate that free radicals are produced during the metabolism of estrone 3,4-quinone in human cells.


Assuntos
Neoplasias da Mama/genética , Dano ao DNA , DNA de Neoplasias/efeitos dos fármacos , Estrenos/toxicidade , Estrogênios/toxicidade , Neoplasias da Mama/metabolismo , Espectroscopia de Ressonância de Spin Eletrônica , Estrenos/metabolismo , Estrogênios/metabolismo , Radicais Livres , Humanos , Peróxido de Hidrogênio/análise , Quinonas/metabolismo , Espécies Reativas de Oxigênio , Frações Subcelulares/efeitos dos fármacos , Células Tumorais Cultivadas
14.
Anal Biochem ; 200(1): 27-30, 1992 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-1595897

RESUMO

A sensitive and facile assay for heme oxygenase (HO) has been developed. The basis of the assay is the detection of [14C]bilirubin formation in a coupled enzyme assay involving HO and biliverdin reductase actions, respectively. Separation of substrate from product is accomplished by thin-layer chromatography with subsequent quantitation by liquid scintillation counting of radioactive material present on chromatograms. As little as 20 micrograms of total cellular protein derived from cells growing in a standard 25-cm2 culture flask is sufficient for detection of HO enzyme activity using this assay. The reaction is inhibited by tin-protoporphyrin (10 microM final concentration), a specific inhibitor of HO. The linearity of the enzyme reaction with respect to incubation time and amount of protein used was established. Comparison of the new HO assay with a spectrophotometric assay was made, and good agreement of the results from both methods was found. The assay described here should facilitate measurements of this important heme-degrading enzyme in tissue culture studies and cases where limited amounts of material are available.


Assuntos
Heme Oxigenase (Desciclizante)/metabolismo , Autorradiografia , Bilirrubina/isolamento & purificação , Neoplasias da Mama/enzimologia , Cromatografia em Camada Fina , Heme/isolamento & purificação , Heme/metabolismo , Heme Oxigenase (Desciclizante)/antagonistas & inibidores , Humanos , Metaloporfirinas/farmacologia , Protoporfirinas/farmacologia , Especificidade por Substrato , Células Tumorais Cultivadas/enzimologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...