Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Mais filtros








Intervalo de ano de publicação
1.
Ann N Y Acad Sci ; 935: 266-74, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11411172

RESUMO

On the surface, higher education has served the nation well. An increasing percentage of adults have attended college. Information is widely and freely available to most citizens; the economy is steady; and technological developments appear to offer ever-increasing improvements in the quality of life. Looking just beneath the surface, though, one realizes that knowledge is fragmented, access to knowledge is not universal, and there are no guarantees that past successes will sustain the future. Thus, it is worth examining how higher learning has contributed to the general prosperity and how education might evolve to address society's evident problems. This essay discusses the core approach of liberal arts in the context of the value--and obstacles--of integration across the traditional disciplines. Critical to the natural sciences is a firm grounding in the central importance of rigorous evidence, while the humanities keep us rooted in the importance of human values. Seeking linkages and connections between these realms is the lively challenge of this conference. The details of curricular design will be of less interest at this juncture than locating the critical issues and discussing how education might serve the goal of unifying knowledge and learning.


Assuntos
Educação/tendências , Aprendizagem , Previsões , Humanos
2.
Cancer Res ; 61(6): 2558-61, 2001 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-11289131

RESUMO

Anthracyclines are, above all, DNA intercalators, which induce genetic damage leading to cell death. However, increasing evidence firmly suggests that the underlying mechanism for anthracycline cytotoxicity is the induction of apoptosis through intracellular-mediated signaling pathways. Whether drug/DNA interaction is necessary for such apoptosis signaling is unknown. We investigated the cellular effects of the anthracyclines daunorubicin (DNR) and doxorubicin (DOX) using the myeloid leukemia cell line U937. By comparing free drug against agarose bead-immobilized drug iDNR and iDOX (which cannot accumulate within the cell), we observed that whereas both free and immobilized anthracyclines were cytotoxic, only the former induced apoptosis; the latter induced necrosis. Indeed, we did not observe ceramide generation, neutral sphingomyelinase activation, poly (ADP-ribose) polymerase cleavage, or other apoptotic events with iDNR or iDOX. However, both free and immobilized drug were similarly capable of triggering nuclear factor kappaB activation. These observations demonstrate that whereas activation of certain cellular signaling pathways can be achieved solely through membrane interaction, apoptosis signaling requires anthracycline internalization. These results also show that the initiation of cell survival pathways (illustrated by nuclear factor kappaB activation) is independent of intracellular drug/target interaction.


Assuntos
Antibióticos Antineoplásicos/toxicidade , Apoptose/efeitos dos fármacos , Daunorrubicina/toxicidade , Doxorrubicina/toxicidade , NF-kappa B/fisiologia , Transdução de Sinais/efeitos dos fármacos , Apoptose/fisiologia , Membrana Celular/efeitos dos fármacos , Ceramidas/biossíntese , Fragmentação do DNA , Humanos , Transdução de Sinais/fisiologia , Células U937
3.
Am J Physiol ; 277(4): L684-93, 1999 10.
Artigo em Inglês | MEDLINE | ID: mdl-10516208

RESUMO

We examined the mechanisms of interaction of crocidolite asbestos fibers with the epidermal growth factor (EGF) receptor (EGFR) and the role of the EGFR-extracellular signal-regulated kinase (ERK) signaling pathway in early-response protooncogene (c-fos/c-jun) expression and apoptosis induced by asbestos in rat pleural mesothelial (RPM) cells. Asbestos fibers, but not the nonfibrous analog riebeckite, abolished binding of EGF to the EGFR. This was not due to a direct interaction of fibers with ligand, inasmuch as binding studies using fibers and EGF in the absence of membranes showed that EGF did not adsorb to the surface of asbestos fibers. Exposure of RPM cells to asbestos caused a greater than twofold increase in steady-state message and protein levels of EGFR (P < 0.05). The tyrphostin AG-1478, which inhibits the tyrosine kinase activity of the EGFR, but not the tyrphostin A-10, which does not affect EGFR activity, significantly ameliorated asbestos-induced increases in mRNA levels of c-fos but not of c-jun. Pretreatment of RPM cells with AG-1478 significantly reduced apoptosis in cells exposed to asbestos. Our findings suggest that asbestos-induced binding to EGFR initiates signaling pathways responsible for increased expression of the protooncogene c-fos and the development of apoptosis. The ability to block asbestos-induced elevations in c-fos mRNA levels and apoptosis by small-molecule inhibitors of EGFR phosphorylation may have therapeutic implications in asbestos-related diseases.


Assuntos
Apoptose/fisiologia , Asbesto Crocidolita/farmacologia , Receptores ErbB/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Animais , Células Cultivadas , Fator de Crescimento Epidérmico/antagonistas & inibidores , Fator de Crescimento Epidérmico/metabolismo , Fator de Crescimento Epidérmico/farmacologia , Células Epiteliais/metabolismo , Receptores ErbB/genética , Receptores ErbB/fisiologia , Regulação da Expressão Gênica/fisiologia , Homeostase/efeitos dos fármacos , Fosforilação , Pleura/citologia , Pleura/metabolismo , Proteínas Proto-Oncogênicas c-fos/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos F344 , Transdução de Sinais/fisiologia
4.
Braz J Med Biol Res ; 32(7): 827-33, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10454740

RESUMO

Resistance to anticancer drugs is a major cause of failure of many therapeutic protocols. A variety of mechanisms have been proposed to explain this phenomenon. The exact mechanism depends upon the drug of interest as well as the tumor type treated. While studying a cell line selected for its resistance to cisplatin we noted that the cells expressed a > 25,000-fold collateral resistance to methotrexate. Given the magnitude of this resistance we elected to investigate this intriguing collateral resistance. From a series of investigations we have identified an alteration in a membrane protein of the resistant cell as compared to the sensitive cells that could be the primary mechanism of resistance. Our studies reviewed here indicate decreased tyrosine phosphorylation of a protein (molecular mass = 66) in the resistant cells, which results in little or no transfer of methotrexate from the medium into the cell. Since this is a relatively novel function for tyrosine phosphorylation, this information may provide insight into possible pharmacological approaches to modify therapeutic regimens by analyzing the status of this protein in tumor samples for a better survival of the cancer patients.


Assuntos
Antineoplásicos/farmacologia , Cisplatino/farmacologia , Leucemia L1210/tratamento farmacológico , Metotrexato/farmacologia , Animais , Antineoplásicos/efeitos adversos , Cisplatino/efeitos adversos , Resistencia a Medicamentos Antineoplásicos , Metotrexato/farmacocinética , Camundongos , Fosforilação , Tirosina/farmacologia
5.
Braz. j. med. biol. res ; 32(7): 827-33, July 1999.
Artigo em Inglês | LILACS | ID: lil-234887

RESUMO

Resistance to anticancer drugs is a major cause of failure of many therapeutic protocols. A variety of mechanisms have been proposed to explain this phenomenon. The exact mechanism depends upon the drug of interest as well as the tumor type treated. While studying a cell line selected for its resistance to cisplatin we noted that the cells expressed a >25,000-fold collateral resistance to methotrexate. Given the magnitude of this resistance we elected to investigate this intriguing collateral resistance. From a series of investigations we have identified an alteration in a membrane protein of the resistant cell as compared to the sensitive cells that could be the primary mechanism of resistance. Our studies reviewed here indicate decreased tyrosine phosphorylation of a protein (molecular mass = 66) in the resistant cells, which results in little or no transfer of methotrexate from the medium into the cell. Since this is a relatively novel function for tyrosine phosphorylation, this information may provide insight into possible pharmacological approaches to modify therapeutic regimens by analyzing the status of this protein in tumor samples for a better survival of the cancer patients


Assuntos
Camundongos , Animais , Antineoplásicos/farmacologia , Cisplatino/farmacologia , Leucemia L1210 , Metotrexato/farmacologia , Antineoplásicos/efeitos adversos , Cisplatino/efeitos adversos , Resistência a Medicamentos , Metotrexato/farmacocinética , Fosforilação , Tirosina/farmacologia
6.
Immunol Cell Biol ; 76(4): 350-6, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9723776

RESUMO

It is demonstrated that methotrexate/cisplatin-sensitive L1210 cells express low levels of major histocompatibility complex (MHC) class II relative to the high levels expressed on methotrexate (MTX)/cisplatin-resistant L1210/DDP cells. L1210 cells express cell-surface Fas, while the L1210/DDP cells express no cell-surface Fas. Expression of costimulatory molecules B7-1/B7-2 and Fas is increased on L1210 cells, but not L1210/DDP, in the presence of methotrexate or trimetrexate (TMTX). Therefore, a component of the mechanism of action of some anti-cancer agents may be to facilitate immune recognition and T cell-directed, Fas-induced cell death. Loss of cell-surface Fas expression and failure of Fas (CD95)-dependent apoptotic death has been observed when cells develop drug resistance. The defect in apoptosis can be overcome by anti-cancer agents or experimental manipulation that induce Fas expression on the drug-resistant cells.


Assuntos
Antígenos CD/biossíntese , Antimetabólitos Antineoplásicos/farmacologia , Antígeno B7-1/biossíntese , Sistema Imunitário/efeitos dos fármacos , Glicoproteínas de Membrana/biossíntese , Receptor fas/biossíntese , Animais , Apoptose , Antígeno B7-2 , Carcinógenos/farmacologia , Linhagem Celular , Relação Dose-Resposta a Droga , Resistência a Medicamentos , Cinética , Metotrexato/farmacologia , Camundongos , Estaurosporina/farmacologia , Trimetrexato/farmacologia
7.
In Vivo ; 12(1): 75-84, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9575429

RESUMO

Glioblastoma multiforme is a malignant primary brain tumor associated with short patient survival in part because of the ability of individual cells to migrate significant distances into brain tissue. Invasion is a difficult process to model, because many such human tumors do not invade immunologically competent animal tissue, tumors grown in animals do not invade human tissue, and relevant human tissue substrates are not easily reproduced. We discuss models for examining invasion in vitro, and in particular review work using the tumor spheroid--fetal rat brain aggregate co-culture model, assessed with confocal microscopy and four-dimensional imaging. Quantitation of invasion in this model is discussed, as well as the invasion-inhibitory properties of tyrosine kinase (TK) inhibitors. The effects of receptor-specific tyrphostins strongly support a dominant role for Epidermal Growth Factor Receptor activation in this process and show that invasion can be effectively inhibited at much lower concentrations of TK inhibitors than is necessary for growth suppression. Inhibition of activation of the purported growth factor receptor second messenger phospholipase C- gamma 1, by pharmacological means and gene transfection, also profoundly inhibits the invasive properties of human glioblastoma and rat C6 glioma cells. We have assessed invasiveness in several human tumor specimens, which may provide information relative to prognosis and recurrence risk. Our data supports the concept of differential control of invasion and proliferation, and points to possible strategies for anti-invasive therapy for glioblastoma multiforme.


Assuntos
Encéfalo/fisiopatologia , Glioblastoma/fisiopatologia , Animais , Encéfalo/embriologia , Humanos , Invasividade Neoplásica , Ratos
8.
Oncol Rep ; 5(2): 419-21, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9468571

RESUMO

We have previously shown that methotrexate (MTX) transport is impaired in murine L1210 cells selected for cisplatin (DDP) resistance (L1210/DDP) and that the decreased MTX uptake may be due to an altered 66 kDa membrane protein. We have further hypothesized that tyrosine phosphorylation is necessary for the function of this protein. To determine the importance of tyrosine phosphorylation we studied the effect of genistein, a tyrosine kinase inhibitor, on methotrexate sensitivity, uptake and tyrosine phosphorylation of the 66 kDa protein in L1210/0 cells. After 5 h of treatment with 50 microM genistein, methotrexate uptake was decreased by nearly 50% and the cells were protected from methotrexate cytotoxicity. Immunoblotting of whole cell lysates with a phosphotyrosine monoclonal antibody demonstrated that genistein treatment decreased phosphorylation of the 66 kDa membrane protein. We concluded that phosphorylation of a 66 kDa protein may be critical for methotrexate transport and that genistein protects L1210/0 cells from methotrexate toxicity.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Inibidores Enzimáticos/farmacologia , Genisteína/farmacologia , Leucemia L1210/tratamento farmacológico , Metotrexato/farmacologia , Animais , Antimetabólitos Antineoplásicos/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos , Leucemia L1210/metabolismo , Metotrexato/metabolismo , Camundongos , Fosforilação , Proteínas Tirosina Quinases/antagonistas & inibidores , Células Tumorais Cultivadas , Tirosina/metabolismo
9.
Neurosurgery ; 40(1): 141-51, 1997 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8971836

RESUMO

OBJECTIVE: Glioblastoma multiforme is a malignant primary brain tumor associated with short patient survival despite aggressive treatment, in part because of its propensity to aggressively infiltrate into brain tissue. Glioblastoma multiforme is also unique because it is the only nonepithelial human tumor for which excessive activation of epidermal growth factor receptor (EGFR) has been consistently linked to tumor growth and patient survival, and EGFR activation promotes glioblastoma multiforme infiltration in vitro. METHODS: Cocultures of human glioblastoma spheroids (derived from three separate patients) and fetal rat brain aggregates were examined for infiltration using confocal microscopy, in the presence of 0 to 100 mumol/L genistein, a tyrosine kinase (TK) inhibitor, and 3 mumol/L tyrphostin A25, a specific EGFR-TK inhibitor. RESULTS: Infiltration (not attachment) was completely inhibited by genistein at 10 mumol/L, the IC20 for monolayer growth inhibition in two cell lines. Tyrphostin A25 at 3 mumol/L (the IC20 for monolayers) reduced invasion in a third cell line from 38.8 +/- 6.1% invasion-hour per hour (n = 5) to 2.9 +/- 1.2% invasion-hour per hour (n = 6) (P = 0.0002, two-tailed t test, 93% inhibition), and from 0.54 +/- 0.065% per hour (slope) to 0.028 +/- 0.018% per hour (P = 0.00001, 95% inhibition). Maximal percent invasion was reduced from 100 +/- 0 to 7.4 +/- 5.6% of the fetal rat brain aggregate. No change was detected in EGFR-associated tyrosine phosphorylation at those doses in monolayers by 32P immunolabeling, consistent with the known effects of low concentrations of TK inhibitors. An increase in expression of wild-type and truncated EGFR was demonstrated by Western blotting. Invasion was equally well inhibited by a monoclonal antibody to the high-affinity ligand binding domain of EGFR and not by antibody to an inactive domain. CONCLUSION: Our observations support the role of EGFR activation as a determinant by which glioblastoma invades normal brain tissue, and we show that invasion can be effectively inhibited at much lower concentrations of TK inhibitors than are necessary for growth suppression.


Assuntos
Neoplasias Encefálicas/patologia , Inibidores Enzimáticos/farmacologia , Receptores ErbB/antagonistas & inibidores , Glioblastoma/patologia , Isoflavonas/farmacologia , Nitrilas/farmacologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Tirfostinas , Animais , Encéfalo/patologia , Divisão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Receptores ErbB/fisiologia , Genisteína , Humanos , Microscopia Confocal , Invasividade Neoplásica , Proteínas Tirosina Quinases/fisiologia , Ratos , Esferoides Celulares/efeitos dos fármacos , Esferoides Celulares/patologia
10.
Cancer Res ; 56(23): 5334-8, 1996 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-8968079

RESUMO

Asbestos fibers are human carcinogens with undefined mechanisms of action. In studies here, we examined signal transduction events induced by asbestos in target cells of mesothelioma and potential cell surface origins for these cascades. Asbestos fibers, but not their nonfibrous analogues, induced protracted phosphorylation of the mitogen-activated protein (MAP) kinases and extracellular signal-regulated kinases (ERK) 1 and 2, and increased kinase activity of ERK2. ERK1 and ERK2 phosphorylation and activity were initiated by addition of exogenous epidermal growth factor (EGF) and transforming growth factor-alpha, but not by isoforms of platelet-derived growth factor or insulin-like growth factor-1 in mesothelial cells. MAP kinase activation by asbestos was attenuated by suramin, which inhibits growth factor receptor interactions, or tyrphostin AG 1478, a specific inhibitor of EGF receptor tyrosine kinase activity (IC50 = 3 nM). Moreover, asbestos caused autophosphorylation of the EGF receptor, an event triggering the ERK cascade. These studies are the first to establish that a MAP kinase signal transduction pathway is initiated after phosphorylation of a peptide growth factor receptor following exposure to asbestos fibers.


Assuntos
Asbesto Crocidolita/farmacologia , Amianto/farmacologia , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Receptores ErbB/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno , Pleura/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Animais , Ativação Enzimática/efeitos dos fármacos , Receptores ErbB/metabolismo , Humanos , Fator de Crescimento Insulin-Like I/farmacologia , Proteína Quinase 1 Ativada por Mitógeno , Proteína Quinase 3 Ativada por Mitógeno , Fosforilação , Fator de Crescimento Derivado de Plaquetas/farmacologia , Pleura/metabolismo , Processamento de Proteína Pós-Traducional , Ratos , Ratos Endogâmicos F344
11.
Biochem Biophys Res Commun ; 226(3): 819-21, 1996 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-8831695

RESUMO

Expression of murine P-glycoprotein (P-gp), encoded by mdrl or mdri3, confers a multidrug resistance phenotype. Higher expression of c-fos and c-jun has also been demonstrated in multidrug resistant human and murine cells. We detected increased expression of c-fos early in the derivation of two series of murine erythroleukemia sublines selected for resistance to vincristine or adriamycin which eventually overexpress mdr3. We speculate that early expression of c-fos prepares cells for overexpression of other genes, such as mdr3, that contribute to the multidrug resistance phenotype.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/biossíntese , Subfamília B de Transportador de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/biossíntese , Doxorrubicina/toxicidade , Resistência a Múltiplos Medicamentos , Proteínas Proto-Oncogênicas c-fos/biossíntese , Vincristina/toxicidade , Animais , Northern Blotting , Linhagem Celular , Expressão Gênica , Genes fos , Genes jun , Humanos , Leucemia Eritroblástica Aguda , Camundongos , Proteínas Proto-Oncogênicas c-jun/biossíntese , Células Tumorais Cultivadas
12.
J Cell Biochem ; 61(1): 9-17, 1996 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-8726350

RESUMO

We report here that the antiestrogen tamoxifen (TAM) induces cell death in human breast cancer cell line MCF-7. We assessed the type of cell death induced by TAM in this breast cancer cell line on the basis of morphological and biochemical characteristics. Dying cells showed morphological characteristics of apoptosis, such as chromatin condensation and nuclear disintegration. DNA isolated from these cells revealed a pattern of distinctive DNA bands on agarose gel. The DNA fragmentation in MCF-7 cells induced by TAM could also be detected by terminal deoxynucleotidyl transferase-mediated dUTP-biotin end labeling. Northern blot hybridization revealed a substantial increase in the amounts of TRPM-2 and TGF-beta 1 mRNAs in MCF-7 cells after treatment with TAM. In contrast, the mRNA level of the estrogen-induced pS2 gene was strongly suppressed. The biological activity of TGF-beta was increased at least fourfold in the media from MCF-7 cells treated with TAM. The results presented in this study suggest that TAM induces apoptosis of MCF-7 cells and it may be mediated by the secretion of active TGF-beta.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Chaperonas Moleculares , Proteínas , Tamoxifeno/farmacologia , Fator de Crescimento Transformador beta/biossíntese , Northern Blotting , Contagem de Células , Cromatografia em Agarose , Clusterina , DNA de Neoplasias/efeitos dos fármacos , Estrogênios/genética , Feminino , Expressão Gênica/efeitos dos fármacos , Glicoproteínas/genética , Humanos , Hibridização In Situ , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Fator de Crescimento Transformador beta/genética , Fator Trefoil-1 , Células Tumorais Cultivadas , Proteínas Supressoras de Tumor
13.
Biochem Pharmacol ; 51(4): 477-82, 1996 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-8619893

RESUMO

Collateral resistance to cisplatin and methotrexate has been reported in several cell lines. A murine leukemia cell line (L1210/DDP) selected for cisplatin resistance also has been shown to be highly resistant to methotrexate. Of the mechanisms proposed for methotrexate resistance, only changes in methotrexate transport into the cells were found in an earlier report. Methotrexate enters mammalian cells via an active transport system. In the present study, we demonstrated that the transport into the cell may be impaired in the resistant cells due to altered tyrosine phosphorylation of a membrane protein with a molecular mass of 66 kDa. This alteration was manifested by altered tyrosine phosphorylation of the 66 kDa protein and may be an underlying modification that renders the cells resistant to methotrexate. These results suggest involvement of tyrosine phosphorylation in folate transport and methotrexate resistant in L1210/DDP cells.


Assuntos
Cisplatino/toxicidade , Resistencia a Medicamentos Antineoplásicos , Leucemia L1210/metabolismo , Proteínas de Membrana/metabolismo , Metotrexato/toxicidade , Fosfoproteínas/metabolismo , Fosfotirosina/metabolismo , Marcadores de Afinidade , Animais , Transporte Biológico Ativo , Western Blotting , Células Clonais , Eletroforese em Gel de Poliacrilamida , Proteínas de Membrana/isolamento & purificação , Metotrexato/metabolismo , Camundongos , Peso Molecular , Fosfoproteínas/isolamento & purificação , Células Tumorais Cultivadas
14.
Cancer Chemother Pharmacol ; 37(4): 337-42, 1996.
Artigo em Inglês | MEDLINE | ID: mdl-8548879

RESUMO

We report a murine leukemia cell variant (L1210/DDP), selected for cisplatin (DDP) resistance, to be cross-resistant to methotrexate (MTX). Cross-resistance of L1210 cells to DDP and MTX has been observed by others, and has also been recorded in P388 murine leukemia and SSC-25 human squamous carcinoma cells. We demonstrated that MTX resistance is not due to dihydrofolate reductase (DHFR) gene amplification, increased DHFR enzyme activity or decreased MTX binding to the target enzyme. Of the mechanisms commonly proposed for MTX resistance, only differences in transport were observed when comparing sensitive (L1210/0) and resistant (L1210/DDP) cells. Our results suggest that MTX resistance in L1210/DDP cells is due to altered methotrexate uptake.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Antineoplásicos/farmacologia , Cisplatino/farmacologia , Leucemia L1210/fisiopatologia , Metotrexato/farmacologia , Animais , Southern Blotting , Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Humanos , Leucemia L1210/tratamento farmacológico , Leucemia L1210/enzimologia , Camundongos , Peptídeo Sintases/metabolismo , Tetra-Hidrofolato Desidrogenase/metabolismo , Fatores de Tempo , Células Tumorais Cultivadas
15.
Cancer Res ; 53(21): 5237-43, 1993 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-8106144

RESUMO

We present studies which suggest that the cytotoxic action of Adriamycin (ADR) may involve intracellular pathways of vesicular transport. The movement of proteins or lipids from the endoplasmic reticulum to the plasma membrane via the Golgi organelle and associated compartments exhibits several temperature-sensitive steps between 15 degrees C and 20 degrees C. In this same temperature range, ADR loses its cytotoxic capacity. Using the inhibitor brefeldin A (BFA), we have investigated the possible role of intravesicular trafficking in the loss of ADR activity and the induction of protection from cytotoxicity at low temperature in L1210 cells. We show here that cells pretreated at 37 degrees C for 2 h with BFA could be protected from a subsequent exposure to ADR. The concentration causing 50% inhibition, determined by cloning in soft agar, was increased approximately 3.5 fold. L1210 cells could also be protected from the topoisomerase II inhibitors etoposide and amsacrine, but to a lesser extent; the concentration causing 50% inhibition for the latter inhibitors were increased 2-fold. Spectrofluorometric analysis of intracellular ADR accumulation revealed that there was no significant difference in the level of ADR in cells with or without BFA pretreatment. In addition, examination of ADR-induced cleavable complex formation by alkaline elution showed no significant difference in the level of DNA strand breaks in cells which had been pretreated with BFA even though there was a large difference in survival. Further examination of the persistence of DNA damage after a period of up to 6 h of repair revealed that cells which were pretreated with BFA removed DNA strand breaks at rates equivalent to those of cells which had received ADR directly. These results suggest that the protective effect induced by brefeldin A does not involve uptake, DNA damage, or repair but instead implicates protein or lipid interactions which may be independent of DNA damage and which may influence critical events that take place after the topoisomerase II-DNA complex has been formed.


Assuntos
Sobrevivência Celular/efeitos dos fármacos , Ciclopentanos/farmacologia , Dano ao DNA , Doxorrubicina/toxicidade , Leucemia L1210/patologia , Amsacrina/toxicidade , Animais , Transporte Biológico , Brefeldina A , Reparo do DNA/efeitos dos fármacos , Doxorrubicina/metabolismo , Sinergismo Farmacológico , Retículo Endoplasmático/metabolismo , Etoposídeo/toxicidade , Complexo de Golgi/metabolismo , Cinética , Leucemia L1210/metabolismo , Camundongos , Microtúbulos/efeitos dos fármacos , Microtúbulos/ultraestrutura , Nocodazol/toxicidade , Paclitaxel/toxicidade , Temperatura , Células Tumorais Cultivadas , Ensaio Tumoral de Célula-Tronco
16.
Biochem Pharmacol ; 46(2): 265-71, 1993 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-8394077

RESUMO

We have examined the activities of two novel aza-anthracene-9,10-diones (aza), 1-aza and 2-aza, in HL-60 human leukemia cell lines containing type II topoisomerases with different sensitivities to inhibition by other intercalating agents. The sensitive line, HL-60, was sensitive to 2-aza but not to 1-aza, whereas the resistant HL-60/AMSA was sensitive to neither agent. Measurements of 1- and 2-aza-induced, topoisomerase II-mediated DNA cross-linking in the cells revealed patterns of resistance and sensitivity that paralleled the results in the cytotoxicity assays. However, measurements of drug-induced topoisomerase II-mediated DNA cross-linking using purified HL-60 and HL-60/AMSA topoisomerase II indicated that both agents could stabilize a covalent complex between DNA and the HL-60 enzyme. HL-60/AMSA topoisomerase II resisted stabilization by either agent. This suggests that the resistance of HL-60 cells to 1-aza is not due to the inability of this drug to inhibit topoisomerase II but rather to another, undefined mechanism.


Assuntos
Antraquinonas/farmacologia , DNA Topoisomerases Tipo II/metabolismo , Substâncias Intercalantes/farmacologia , Isoquinolinas/farmacologia , Quinolonas , DNA/metabolismo , Resistência a Medicamentos , Humanos , Leucemia/enzimologia , Inibidores da Topoisomerase II , Células Tumorais Cultivadas/efeitos dos fármacos
17.
Int J Cancer ; 54(2): 302-8, 1993 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-8098016

RESUMO

KB-A1 and KB-A10 are 2 multi-drug-resistant cell lines which are 100- and 1,000-fold resistant to Adriamycin, respectively. We have examined the expression of P-glycoprotein at the molecular and cellular levels in these human carcinoma cells. Both MDR cell lines, when compared to the parental KB-3-1, show characteristic increases in mdr 1 gene copy number, an increase in mdr 1 mRNA expression, a corresponding increase in transcription rate and a consequent over-expression of P-glycoprotein. However, the more highly resistant KB-A10 cells have a lower gene copy number, express less mdr 1 mRNA and contain less P-glycoprotein than the A1 cell line. To determine whether higher levels of cellular resistance were attributable to enhanced efficacy of P-glycoprotein or to other cellular regulatory mechanisms, we examined other major cellular properties known to be associated with the mdr phenotype. Both the KB-A1 and KB-A10 lines exhibit similar increases in protein kinase C activity as compared to the drug-sensitive parent. In addition, neither glutathione-S-transferase nor topoisomerase II activities account for enhanced resistance of the KB-A10 cells. The above observations are contrary to the premise that the level of drug resistance is necessarily proportional to expression of P-glycoprotein or to other common factors thought to participate in drug insensitivity; consequently, new mechanisms of resistance must be in operation in these cells.


Assuntos
Resistência a Medicamentos , Células KB/efeitos dos fármacos , Glicoproteínas de Membrana/metabolismo , Membro 1 da Subfamília B de Cassetes de Ligação de ATP , Western Blotting , DNA Topoisomerases Tipo II/metabolismo , Doxorrubicina/farmacologia , Expressão Gênica , Glutationa Transferase/metabolismo , Humanos , Técnicas In Vitro , Células KB/metabolismo , Glicoproteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Fosfoproteínas/metabolismo , Fosforilação , Proteína Quinase C/metabolismo
18.
Cancer Res ; 52(15): 4135-8, 1992 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-1638527

RESUMO

Adriamycin is a cytotoxic drug which has enjoyed considerable success in the treatment of cancer. This agent has a bewildering variety of biological effects both within and on the surface of cells exposed to drug, and it has proved difficult to unambiguously assign a single mechanism of action. In this report we are able to separate intracellular and extracellular actions by taking advantage of the complete lack of Adriamycin-induced cytotoxicity at low temperature. For example, cells exposed to 100 microM Adriamycin at 0 degree C are not killed by the drug, even though this concentration is orders of magnitude higher than the concentration needed to cause 100% cell death at 37 degrees C. If cells exposed to 100 microM Adriamycin at 0 degree C are shifted to fresh drug-free medium at 37 degrees C, there is a time-dependent decrease in survival. However, if the drug-free medium contains calf thymus DNA (1.5 mg/ml) to act as a reservoir for Adriamycin binding of effluxed drug, there is no ensuing cytotoxicity. Thus, the results show that no matter how much drug is present inside the cell, there must also be extracellular drug available for membrane interaction in order to initiate nuclear DNA damage and the cytotoxic cascade.


Assuntos
Morte Celular/efeitos dos fármacos , Dano ao DNA , Doxorrubicina/farmacologia , Animais , Meios de Cultura , Técnicas de Cultura/métodos , Cinética , Leucemia L1210 , Camundongos , Temperatura , Fatores de Tempo , Células Tumorais Cultivadas
19.
Mol Pharmacol ; 42(1): 69-74, 1992 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-1353251

RESUMO

In both mouse sarcoma 180 and human KB cells selected for the multiple drug resistance (MDR) phenotype, there is an elevation in the steady state mRNA level of c-fos. There is no detectable gene amplification for c-fos, nor is there any significant change in the rate of mRNA transcription or degradation, suggesting that other factors are responsible for the increased expression level in resistance. Cells selected for resistance to methotrexate, a drug not in the MDR group, do not have an increase in c-fos mRNA expression. When drug-sensitive cells are exposed for 30 min to an ED50 concentration of vinblastine, Adriamycin, colchicine, or VP-16, but not to methotrexate or cisplatin, there is a 3-6-fold induction in the level of c-fos message. Because the former drugs are members of the MDR class and the latter are not, the results are consistent with the hypothesis that induction of c-fos by low levels of cytotoxic drugs may be an early event in the acquisition of the MDR phenotype. If this were the case, then c-fos would be expected to act in concert with c-jun to control transcription by binding to a specific DNA regulatory site. Consistent with this explanation is the existence of an AP-1 sequence in the promotor region for the P-glycoprotein gene (mdr1), as well as the fact that c-jun is also overexpressed in MDR cells.


Assuntos
Resistência a Medicamentos/genética , Genes fos , Membro 1 da Subfamília B de Cassetes de Ligação de ATP , Animais , Antineoplásicos/farmacologia , Northern Blotting , Células Cultivadas , Regulação Neoplásica da Expressão Gênica , Genes jun , Humanos , Glicoproteínas de Membrana/genética , Camundongos , Fenótipo , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Transcrição Gênica , Células Tumorais Cultivadas
20.
Pharmacol Ther ; 49(3): 293-309, 1991.
Artigo em Inglês | MEDLINE | ID: mdl-2052627

RESUMO

Adriamycin has a vast range of reported actions on the structural and functional properties of cells. This review summarizes the literature on the ability of the drug to modulate the cell surface membrane and attempts to address the question of how such actions could be linked to cytotoxicity. In addition, we consider the use of polymer immobilization of adriamycin to separate intracellular from plasma membrane effects of the drug, and show how this approach has been helpful in interpreting the pharmacology of adriamycin. Finally, a range of biophysical and spectroscopic approaches to defining the molecular details of adriamycin-bilayer interactions is surveyed, and the results used to discuss a model for how this antineoplastic agent binds to membranes.


Assuntos
Doxorrubicina/farmacologia , Membrana Celular/efeitos dos fármacos , Doxorrubicina/farmacocinética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA