Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Chromosoma ; 110(2): 65-74, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11453556

RESUMO

The inner centromere protein (INCENP), which has previously been described in chicken, frog and mouse, is required for correct chromosome segregation and cytokinesis. We have identified the human INCENP gene by library screening and reverse transcription-polymerase chain reaction (RT-PCR) and localized it to chromosomal region 11q12. HsINCENP is a single-copy gene that consists of 17 exons and covers 25 kb of genomic DNA. The gene is expressed at highest levels in the colon, testis and prostate, consistent with its likely role in cell proliferation. HsINCENP encodes a highly basic protein of 915 amino acids that localizes to metaphase chromosomes and to the mitotic spindle and equatorial cortex at anaphase. Recently we showed that INCENP is stockpiled in a complex with the Aurora-B/XAIRK2 kinase in Xenopus eggs. Here we demonstrate that, consistent with such an interaction, the two proteins colocalize on human metaphase chromosomes. Levels of Aurora-B are increased in several human cancers, and we show here that HsINCENP protein levels are also significantly increased in several colorectal cancer cell lines.


Assuntos
Centrômero/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Cromossomos Humanos/metabolismo , Neoplasias do Colo/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Sequência de Aminoácidos , Animais , Aurora Quinase B , Aurora Quinases , Southern Blotting , Células Cultivadas , Proteínas Cromossômicas não Histona/genética , Cromossomos Humanos/ultraestrutura , Clonagem Molecular , Fibroblastos/citologia , Fibroblastos/metabolismo , Imunofluorescência , Humanos , Metáfase , Camundongos , Dados de Sequência Molecular , Proteínas Serina-Treonina Quinases/genética , RNA Mensageiro/metabolismo , Homologia de Sequência de Aminoácidos , Xenopus laevis
2.
J Clin Invest ; 106(9): 1127-37, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11067865

RESUMO

TNF-alpha-induced apoptosis is thought to involve mediators from acidic vesicles. Cathepsin B (cat B), a lysosomal cysteine protease, has recently been implicated in apoptosis. To determine whether cat B contributes to TNF-alpha-induced apoptosis, we exposed mouse hepatocytes to the cytokine in vitro and in vivo. Isolated hepatocytes treated with TNF-alpha in the presence of the transcription inhibitor actinomycin D (AcD) accumulated cat B in their cytosol. Further experiments using cell-free systems indicated that caspase-8 caused release of active cat B from purified lysosomes and that cat B, in turn, increased cytosol-induced release of cytochrome c from mitochondria. Consistent with these observations, the ability of TNF-alpha/AcD to induce mitochondrial release of cytochrome c, caspase activation, and apoptosis of isolated hepatocytes was markedly diminished in cells from CatB(-/-) mice. Deletion of the CatB gene resulted in diminished liver injury and enhanced survival after treatment in vivo with TNF-alpha and an adenovirus construct expressing the IkappaB superrepressor. Collectively, these observations suggest that caspase-mediated release of cat B from lysosomes enhances mitochondrial release of cytochrome c and subsequent caspase activation in TNF-alpha-treated hepatocytes.


Assuntos
Catepsina B/metabolismo , Grupo dos Citocromos c/metabolismo , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Animais , Apoptose/efeitos dos fármacos , Transporte Biológico Ativo/efeitos dos fármacos , Caspases/metabolismo , Catepsina B/deficiência , Catepsina B/genética , Citosol/metabolismo , Dactinomicina/farmacologia , Ativação Enzimática/efeitos dos fármacos , Hepatócitos/citologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Camundongos , Camundongos Knockout , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Modelos Biológicos , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo
3.
Blood ; 96(12): 3922-31, 2000 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-11090079

RESUMO

Recent studies have suggested that variations in levels of caspases, a family of intracellular cysteine proteases, can profoundly affect the ability of cells to undergo apoptosis. In this study, immunoblotting was used to examine levels of apoptotic protease activating factor-1 (Apaf-1) and procaspases-2, -3, -7, -8, and -9 in bone marrow samples (at least 80% leukemia) harvested before chemotherapy from adults with newly diagnosed acute myelogenous leukemia (AML, 42 patients) and acute lymphocytic leukemia (ALL, 18 patients). Levels of each of these polypeptides varied over a more than 10-fold range between specimens. In AML samples, expression of procaspase-2 correlated with levels of Apaf-1 (R(s) = 0.52, P <.02), procaspase-3 (R(s) = 0.56, P <.006) and procaspase-8 (R(s) = 0.64, P <.002). In ALL samples, expression of procaspases-7 and -9 was highly correlated (R(s) = 0.90, P <.003). Levels of these polypeptides did not correlate with prognostic factors or response to induction chemotherapy. In further studies, 16 paired samples (13 AML, 3 ALL), the first harvested before induction therapy and the second harvested at the time of leukemia regrowth, were also examined. There were no systematic alterations in levels of Apaf-1 or procaspases at relapse compared with diagnosis. These results indicate that levels of initiator caspases vary widely among different leukemia specimens but cast doubt on the hypothesis that this variation is a major determinant of drug sensitivity for acute leukemia in the clinical setting. (Blood. 2000;96:3922-3931)


Assuntos
Caspases/metabolismo , Precursores Enzimáticos/metabolismo , Leucemia/diagnóstico , Doença Aguda , Adolescente , Adulto , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Fator Apoptótico 1 Ativador de Proteases , Biomarcadores Tumorais/imunologia , Biomarcadores Tumorais/metabolismo , Células da Medula Óssea/química , Células da Medula Óssea/enzimologia , Caspases/efeitos dos fármacos , Caspases/imunologia , Estudos de Coortes , Precursores Enzimáticos/efeitos dos fármacos , Precursores Enzimáticos/imunologia , Células HL-60 , Humanos , Immunoblotting , Leucemia/metabolismo , Leucemia/terapia , Leucemia Mieloide/diagnóstico , Leucemia Mieloide/metabolismo , Leucemia Mieloide/terapia , Pessoa de Meia-Idade , Leucemia-Linfoma Linfoblástico de Células Precursoras/diagnóstico , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Prognóstico , Proteínas/efeitos dos fármacos , Proteínas/imunologia , Proteínas/metabolismo
4.
Clin Cancer Res ; 6(6): 2318-25, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10873082

RESUMO

Farnesyltransferase inhibitors (FTIs) were developed to target abnormal signaling pathways that are commonly activated in neoplastic cells. Five FTIs have recently undergone Phase I testing; and two are currently in Phase II clinical trials. As part of the development of these agents, there has been interest in determining their cellular effects in the clinical setting. Several approaches have been proposed, including measurement of FT enzymatic activity, evaluation of the processing of FT polypeptide substrates, and assessment of the accumulation of p21waf1. In the present study, a number of these assays have been compared in four cultured human neoplastic cell lines of different histology (A549, HCT116, BxPC-3, and MCF-7) after treatment with the nonpeptidomimetic FTI SCH66336 and the peptidomimetic inhibitor FTI-277. Immunoblotting studies failed to demonstrate a mobility shift in ras proteins or increased accumulation of p21waf1 after treatment with these agents. In contrast, drug-induced increases in the slower migrating, unprocessed species of the chaperone protein HDJ-2 and the intranuclear intermediate filament protein lamin A were detected in all four cell lines after treatment with either agent. Unprocessed forms of both polypeptides accumulated in noncycling as well as cycling cells. The precursor peptide that is present in prelamin A but absent from mature lamin A could be readily detected by immunohistochemistry in noncycling cells with a peptide-specific antiserum. Our results indicate that unprocessed HDJ-2 and prelamin A should be suitable markers of FT inhibition in clinical samples.


Assuntos
Alquil e Aril Transferases/antagonistas & inibidores , Antineoplásicos/farmacologia , Inibidores Enzimáticos/farmacologia , Metionina/análogos & derivados , Metionina/farmacologia , Piperidinas/farmacologia , Piridinas/farmacologia , Sequência de Aminoácidos , Biomarcadores Tumorais , Proteínas de Transporte/metabolismo , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/biossíntese , Relação Dose-Resposta a Droga , Eletroforese em Gel de Poliacrilamida , Farnesiltranstransferase , Proteínas de Choque Térmico HSP40 , Proteínas de Choque Térmico/metabolismo , Humanos , Immunoblotting , Imuno-Histoquímica , Concentração Inibidora 50 , Lamina Tipo A , Laminas , Dados de Sequência Molecular , Proteínas Nucleares/metabolismo , Peptídeos/química , Precursores de Proteínas/metabolismo , Prenilação de Proteína , Transdução de Sinais/efeitos dos fármacos , Transfecção , Células Tumorais Cultivadas
5.
Cancer Res ; 60(9): 2419-28, 2000 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-10811119

RESUMO

Flavopiridol, the first potent cyclin-dependent kinase inhibitor to enter clinical trials, was recently found to be cytotoxic to noncycling cells. The present studies were performed to examine the hypothesis that flavopiridol, like several other antineoplastic agents that kill noncycling cells, might also interact with DNA. Consistent with this possibility, treatment of A549 human lung cancer cells with clinically achievable concentrations of flavopiridol resulted in rapid elevations of the DNA damage-responsive protein p53. In further studies, the binding of flavopiridol to DNA was examined in vitro by four independent techniques. Absorption spectroscopy revealed that addition of DNA to aqueous flavopiridol solutions resulted in a red shift of the flavopiridol lambda(max) from 311 to 344 nm, demonstrating an isosbestic point typical of changes seen with DNA-binding compounds. Reverse-phase high-performance liquid chromatography demonstrated that flavopiridol binds to genomic DNA to a similar extent as ethidium bromide and Hoechst 33258. Nuclear magnetic resonance spectroscopy revealed that DNA caused extreme broadening of flavopiridol 1H nuclear magnetic resonance signals that could be reversed by addition of ethidium bromide or by DNA melting, suggesting that flavopiridol binds to (and likely intercalates into) duplex DNA. Equilibrium dialysis demonstrated that the equilibrium dissociation constant of the flavopiridol-DNA complex (5.4+/-3.4 x 10(-4) M) was in the same range observed for binding of the intercalators doxorubicin and pyrazoloacridine to DNA. Molecular modeling confirmed the feasibility of flavopiridol intercalation into DNA and analysis of the effects of flavopiridol in the National Cancer Institute tumor cell line panel using the COMPARE algorithm demonstrated that flavopiridol most closely resembles cytotoxic antineoplastic intercalators. Collectively, these data suggest that DNA might be a second target of flavopiridol, providing a potential explanation for the ability of this agent to kill noncycling cancer cells.


Assuntos
Antineoplásicos/metabolismo , DNA/metabolismo , Inibidores Enzimáticos/metabolismo , Flavonoides/metabolismo , Piperidinas/metabolismo , Acridinas/farmacologia , Animais , Apigenina , Bovinos , Cromatografia Líquida de Alta Pressão , Quinases Ciclina-Dependentes/antagonistas & inibidores , DNA/efeitos dos fármacos , DNA/efeitos da radiação , DNA Topoisomerases Tipo I/metabolismo , Relação Dose-Resposta a Droga , Relação Dose-Resposta à Radiação , Inibidores Enzimáticos/farmacologia , Etídio/farmacologia , Flavonoides/farmacologia , Corantes Fluorescentes/farmacologia , Humanos , Immunoblotting , Substâncias Intercalantes/farmacologia , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Piperidinas/farmacologia , Pirazóis/farmacologia , RNA/efeitos dos fármacos , RNA/metabolismo , Fatores de Tempo , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/biossíntese
6.
Cancer Res ; 60(7): 1871-7, 2000 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-10766174

RESUMO

Farnesyl protein transferase (FT), an enzyme that catalyzes the first step in the posttranslational modification of ras and a number of other polypeptides, has emerged as an important target for the development of anticancer agents. SCH66336 is one of the first FT inhibitors to undergo clinical testing. We report a Phase I trial to assess the maximum tolerated dose, toxicities, and biological effectiveness of SCH66336 in inhibiting FT in vivo. Twenty patients with solid tumors received 92 courses of escalating SCH66336 doses given orally twice a day (b.i.d.) for 7 days out of every 3 weeks. Gastrointestinal toxicity (nausea, vomiting, and diarrhea) and fatigue were dose-limiting at 400 mg of SCH66336 b.i.d. Moderate reversible renal insufficiency, secondary to dehydration from gastrointestinal toxicity, was also seen. Inhibition of prelamin A farnesylation in buccal mucosa cells of patients treated with SCH66336 was demonstrated, confirming that SCH66336 inhibits protein farnesylation in vivo. One partial response was observed in a patient with previously treated metastatic non-small cell lung cancer, who remained on study for 14 months. This study not only establishes the dose for future testing on this schedule (350 mg b.i.d.) but also provides the first evidence of successful inhibition of FT in the clinical setting and the first hint of clinical activity for this class of agents.


Assuntos
Alquil e Aril Transferases/antagonistas & inibidores , Antineoplásicos/efeitos adversos , Neoplasias/tratamento farmacológico , Piperidinas/efeitos adversos , Piridinas/efeitos adversos , Adulto , Idoso , Idoso de 80 Anos ou mais , Antineoplásicos/administração & dosagem , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/efeitos adversos , Farnesiltranstransferase , Feminino , Humanos , Lamina Tipo A , Laminas , Masculino , Pessoa de Meia-Idade , Mucosa Bucal/patologia , Proteínas Nucleares/análise , Piperidinas/administração & dosagem , Precursores de Proteínas/análise , Piridinas/administração & dosagem
7.
Mol Pharmacol ; 57(3): 529-38, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10692493

RESUMO

The present study was undertaken to examine the mechanistic basis for the recent observation that the pyridine nucleotide derivative 6-aminonicotinamide (6AN, NSC 21206) enhances the accumulation and resulting cytotoxicity of cisplatin in a variety of tumor cell lines. When A549 lung cancer cells or K562 leukemia cells were treated with 62.5 microM 6AN for 21 h and then pulse-labeled with [(35)S]methionine for 1 h, increased labeling of five polypeptides, one of which corresponded to a M(r) approximately 78,000 glucose-regulated protein (GRP78), was observed. Two subsequent observations, however, suggested that up-regulation of these polypeptides was unlikely to explain the interaction between 6AN and cisplatin: 1) the concentration of 6AN required to induce GRP78 was 4-fold higher than the dose required to sensitize cells to cisplatin; and 2) simultaneous treatment of cells with 6AN and cycloheximide prevented the increase in GRP78 but not the sensitizing effect of 6AN. On the contrary, treatment with the protein synthesis inhibitors cycloheximide, anisomycin, or puromycin as well as prolonged exposure to the RNA synthesis inhibitor actinomycin D mimicked the biochemical modulating effects of 6AN on cisplatin action. Conversely, 6AN inhibited protein synthesis, whereas 18 6AN analogs that failed to enhance Pt-DNA adducts and cisplatin cytotoxicity failed to inhibit protein synthesis. These observations are consistent with a model in which 6AN and other inhibitors of protein synthesis act as modulating agents by increasing cisplatin accumulation, thereby enhancing the formation of Pt-DNA adducts and subsequent cisplatin-induced cell death.


Assuntos
6-Aminonicotinamida/farmacologia , Antineoplásicos/farmacologia , Cisplatino/farmacologia , Adutos de DNA/efeitos dos fármacos , Compostos Organoplatínicos , Inibidores da Síntese de Proteínas/farmacologia , Interações Medicamentosas , Eletroforese em Gel de Poliacrilamida , Chaperona BiP do Retículo Endoplasmático , Humanos , Concentração de Íons de Hidrogênio , Peptídeos/metabolismo , Células Tumorais Cultivadas , Ensaio Tumoral de Célula-Tronco
8.
Clin Cancer Res ; 6(1): 237-49, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10656455

RESUMO

The tyrphostin AG957 (NSC 654705) inhibits p210bcr/abl, the transforming kinase responsible for most cases of chronic myelogenous leukemia (CML). The present studies were performed to determine the fate of AG957-treated cells and assess the selectivity of AG957 for CML myeloid progenitors. When K562 cells (derived from a patient with blast crisis CML) were treated with AG957, dose- and time-dependent p210bc/abl down-regulation was followed by mitochondrial release of cytochrome c, activation of caspase-9 and caspase-3, and apoptotic morphological changes. These apoptotic changes were inhibited by transfection with cDNA encoding dominant negative caspase-9 but not dominant-negative FADD or blocking anti-Fas antibodies. In additional experiments, a 24-h AG957 exposure caused dose-dependent inhibition of K562 colony formation in soft agar. To extend these studies to clinical samples of CML, peripheral blood mononuclear cells from 10 chronic phase CML patients and normal controls were assayed for the growth of hematopoietic colonies in vitro in the presence of increasing concentrations of AG957. These assays demonstrated selectivity of AG957 for CML progenitors, with median IC50s (CML versus normal) of 7.3 versus >20 microM AG957 in granulocyte colony-forming cells (P < 0.001), 5.3 versus >20 microM in granulocyte/macrophage colony-forming cells (P < 0.05), and 15.5 versus > 20 microM in erythroid colony-forming cells (P > 0.05). The adamantyl ester of AG957 (NSC 680410) down-regulated p210bcr/abl in K562 cells and inhibited granulocyte colony formation in CML specimens at lower concentrations without enhanced toxicity in normal progenitors. These observations not only demonstrate that AG957-induced p210bcr/abl down-regulation is followed by activation of the cytochrome c/Apaf-1/caspase-9 pathway but also indicate that this class of kinase inhibitor exhibits selectivity worthy of further evaluation.


Assuntos
Adamantano/análogos & derivados , Inibidores Enzimáticos/toxicidade , Proteínas de Fusão bcr-abl/antagonistas & inibidores , Hidroquinonas/toxicidade , Tirfostinas/toxicidade , Adamantano/toxicidade , Apoptose/efeitos dos fármacos , Caspase 9 , Caspases/metabolismo , Divisão Celular/efeitos dos fármacos , Proteínas de Fusão bcr-abl/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Células K562 , Cinética , Leucemia Mielogênica Crônica BCR-ABL Positiva , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Recombinantes de Fusão/metabolismo , Transfecção , Ensaio Tumoral de Célula-Tronco
9.
Clin Cancer Res ; 6(2): 661-70, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10690552

RESUMO

Flavopiridol, the first inhibitor of cyclin-dependent kinases to enter clinical trials, has shown promising antineoplastic activity and is currently undergoing Phase II testing. Little is known about mechanisms of resistance to this agent. In the present study, we have characterized an ovarian carcinoma cell line [OV202 high passage (hp)] that spontaneously developed drug resistance upon prolonged passage in tissue culture. Standard cytogenetic analysis and spectral karyotyping revealed that OV202 hp and the parental low passage line OV202 shared several marker chromosomes, confirming the relatedness of these cell lines. Immunoblotting demonstrated that OV202 and OV202 hp contained similar levels of a variety of polypeptides involved in cell cycle regulation, including cyclin-dependent kinases 2 and 4; cyclins A, D1, and E; and proliferating cell nuclear antigen. Despite these similarities, OV202 hp was resistant to flavopiridol and cisplatin, with increases of 5- and 3-fold, respectively, in the mean drug concentrations required to inhibit colony formation by 90%. In contrast, OV202 hp and OV202 displayed indistinguishable sensitivities to oxaliplatin, paclitaxel, topotecan, 1,3-bis(2-chloroethyl)-1-nitrosourea, etoposide, doxorubicin, vincristine, and 5-fluorouracil, suggesting that the spontaneously acquired resistance was not attributable to altered P-glycoprotein levels or a general failure to engage the cell death machinery. After incubation with cisplatin, whole cell platinum and platinum-DNA adducts measured using mass spectrometry were lower in OV202 hp cells than OV202 cells. Similarly, after flavopiridol exposure, whole cell flavopiridol concentrations measured by a newly developed high performance liquid chromatography assay were lower in OV202 hp cells. These data are consistent with the hypothesis that acquisition of spontaneous resistance to flavopiridol and cisplatin in OV202 hp cells is due, at least in part, to reduced accumulation of the respective drugs. These observations not only provide the first characterization of a flavopiridol-resistant cell line but also raise the possibility that alterations in drug accumulation might be important in determining sensitivity to this agent.


Assuntos
Antineoplásicos/toxicidade , Aberrações Cromossômicas , Cisplatino/toxicidade , Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Flavonoides/toxicidade , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Piperidinas/toxicidade , Carmustina/toxicidade , Mapeamento Cromossômico , Cisplatino/farmacocinética , Adutos de DNA/análise , Feminino , Humanos , Cariotipagem , Compostos Organoplatínicos/farmacocinética , Compostos Organoplatínicos/toxicidade , Oxaliplatina , Células Tumorais Cultivadas
10.
Clin Cancer Res ; 6(2): 681-92, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10690554

RESUMO

Phenylacetate (PA) and phenylbutyrate (PB) are aromatic fatty acids that are presently undergoing evaluation as potential antineoplastic agents. In vitro, PA and PB cause differentiation or growth inhibition of malignant cells. Clinical trials of these drugs as single agents indicate that they are not myelosuppressive; therefore, combinations with other chemotherapy agents may be possible. The goals of this study were to determine whether PA and PB (a) are cytotoxic to malignant B cells from patients with non-Hodgkin's lymphoma and B-cell chronic lymphocytic leukemia and (b) exhibit additive or synergistic induction of apoptosis when administered to myeloma cell lines in combination with conventional drugs. In the clinical specimens, cytotoxicity was measured by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, and percent apoptosis was measured using 7-aminoactinomycin D and flow cytometry. Viability was decreased by > 50% in 7% (1/15) of non-Hodgkin's lymphoma samples treated with 5 mM PA, 27% treated with 1 mM PB, and 60% treated with 2 mM PB. Likewise, viability was decreased by > 50% in 44% (4/9) of chronic lymphocytic leukemia samples treated with 5 mM PA, 67% treated with 1 mM PB, and 100% treated with 2 mM PB. Studies in the myeloma cell lines demonstrated that PB treatment induced activation of caspases 3, 7, and 9 accompanied by cleavage of their substrates and internucleosomal DNA degradation. Combinations of PA or PB with conventional drugs (cytarabine, topotecan, doxorubicin, etoposide, chlorambucil, melphalan, fludarabine, carboplatin, and cisplatin) were examined for synergism (combination index < 1 in median effect analysis) in inducing apoptosis of both the MY5 and 8226 human myeloma cell lines. At concentrations that killed > 50% of cells, most combinations were additive; however, PB was synergistic with cytarabine, etoposide, and topotecan, with the combination index < 1 at each of the 50, 75, and 95% apoptosis levels. These observations indicate that PA and PB can induce apoptosis in malignant B cells and enhance the cytotoxicity of agents used in the treatment of these malignancies.


Assuntos
Antineoplásicos/toxicidade , Apoptose/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Fenilacetatos/toxicidade , Fenilbutiratos/toxicidade , Interações Medicamentosas , Sinergismo Farmacológico , Humanos , Leucemia Linfocítica Crônica de Células B , Linfoma de Células B , Linfoma não Hodgkin , Mieloma Múltiplo , Células Tumorais Cultivadas
11.
Biochem Pharmacol ; 58(6): 1057-66, 1999 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-10509758

RESUMO

The pyridine nucleotide 6-aminonicotinamide (6AN) was shown recently to sensitize a number of human tumor cell lines to cisplatin in vitro. The present studies were undertaken to compare the drug concentrations and length of exposure required for this sensitization in vitro with the drug exposure that could be achieved in mice in vivo. Human K562 leukemia cells and A549 lung cancer cells were incubated with 6AN for various lengths of time, exposed to cisplatin for 1-2 hr, and assayed for Pt-DNA adducts as well as the ability to form colonies. K562 cells displayed progressive increases in Pt-DNA adducts and cisplatin sensitivity during the first 10 hr of 6AN exposure. An 18-hr 6AN exposure was likewise more effective than a 6-hr 6AN exposure in sensitizing A549 cells to cisplatin. HPLC analysis of 6AN and its metabolite, 6-amino-NAD+, permitted assessment of exposures achieved in vivo after i.v. administration of 10 mg/kg of 6AN to CD2F1 mice. 6AN reached peak serum concentrations of 80-90 microM and was cleared rapidly, with T1/2alpha and T1/2beta values of 7.4 and 31.3 min, respectively. Bioavailability was 80-100% with identical plasma pharmacokinetics after i.p. administration. At least 25% of the 6AN was excreted unchanged in the urine. The metabolite 6-amino-NAD+ was detected in perchloric acid extracts of brain, liver, kidney, and spleen, but not in serum. Efforts to prolong systemic 6AN exposure by administering multiple i.p. doses or using osmotic pumps resulted in lethal toxicity. These results demonstrated that 6AN exposures required to sensitize tumor cells to cisplatin in vitro are difficult to achieve in vivo.


Assuntos
6-Aminonicotinamida/farmacocinética , 6-Aminonicotinamida/administração & dosagem , 6-Aminonicotinamida/metabolismo , 6-Aminonicotinamida/farmacologia , Animais , Antineoplásicos/farmacologia , Divisão Celular/efeitos dos fármacos , Cisplatino/farmacologia , Interações Medicamentosas , Humanos , Células K562 , Camundongos , NAD/análogos & derivados , NAD/farmacocinética , Distribuição Tecidual , Células Tumorais Cultivadas
12.
J Biol Chem ; 274(32): 22635-45, 1999 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-10428844

RESUMO

The present studies compared caspase activation under cell-free conditions in vitro and in etoposide-treated HL-60 leukemia cells in situ. Immunoblotting revealed that incubation of HL-60 cytosol at 30 degrees C in the presence of cytochrome c and ATP (or dATP) resulted in activation of procaspases-3, -6, and -7 but not -2 and -8. Although similar selectivity was observed in intact cells, affinity labeling revealed that the active caspase species generated in vitro and in situ differed in charge and abundance. ATP and dATP levels in intact HL-60 cells were higher than required for caspase activation in vitro and did not change before caspase activation in situ. Replacement of ATP with the poorly hydrolyzable analogs 5'-adenylyl methylenediphosphate, 5'-adenylyl imidodiphosphate, or 5'-adenylyl-O-(3-thiotriphos-phate) slowed caspase activation in vitro, suggesting that ATP hydrolysis is required. Caspase activation in vitro was insensitive to phosphatase and kinase inhibitors (okadaic acid, staurosporine, and genistein) but was inhibited by Zn(2+), aurintricarboxylic acid, and various protease inhibitors, including 3,4-dichloroisocoumarin, N(alpha)-p-tosyl-L-phenylalanine chloromethyl ketone, N(alpha)-p-tosyl-L-lysine chloromethyl ketone, and N-(N(alpha)-benzyloxycarbonylphenylalanyl)alanine fluoromethyl ketone, each of which inhibited recombinant caspases-3, -6, -7, and -9. Experiments with anti-neoepitope antiserum confirmed that these agents inhibited caspase-9 activation. Collectively, these results suggest that caspase-9 activation requires nucleotide hydrolysis and is inhibited by agents previously thought to affect apoptosis by other means.


Assuntos
Apoptose/fisiologia , Caspases/metabolismo , Citosol/metabolismo , Processamento de Proteína Pós-Traducional , Trifosfato de Adenosina/farmacologia , Ácido Aurintricarboxílico/farmacologia , Inibidores de Caspase , Sistema Livre de Células , Grupo dos Citocromos c/farmacologia , Ativação Enzimática , Etoposídeo/farmacologia , Células HL-60 , Humanos , Fosfoproteínas Fosfatases/antagonistas & inibidores , Inibidores de Proteases/farmacologia , Inibidores de Proteínas Quinases , Proteínas Recombinantes/metabolismo
13.
J Biol Chem ; 274(7): 4335-40, 1999 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-9933635

RESUMO

Previous studies have demonstrated that topoisomerase I is cleaved late during apoptosis, but have not identified the proteases responsible or examined the functional consequences of this cleavage. Here, we have shown that treatment of purified topoisomerase I with caspase-3 resulted in cleavage at DDVD146 downward arrowY and EEED170 downward arrowG, whereas treatment with caspase-6 resulted in cleavage at PEDD123 downward arrowG and EEED170 downward arrowG. After treatment of Jurkat T lymphocytic leukemia cells with anti-Fas antibody or A549 lung cancer cells with topotecan, etoposide, or paclitaxel, the topoisomerase I fragment comigrated with the product that resulted from caspase-3 cleavage at DDVD146 downward arrowY. In contrast, two discrete topoisomerase I fragments that appeared to result from cleavage at DDVD146 downward arrowY and EEED170 downward arrowG were observed after treatment of MDA-MB-468 breast cancer cells with paclitaxel. Topoisomerase I cleavage did not occur in apoptotic MCF-7 cells, which lack caspase-3. Cell fractionation and band depletion studies with the topoisomerase I poison topotecan revealed that the topoisomerase I fragment remains in proximity to the chromatin and retains the ability to bind to and cleave DNA. These observations indicate that topoisomerase I is a substrate of caspase-3 and possibly caspase-6, but is cleaved at sequences that differ from those ordinarily preferred by these enzymes, thereby providing a potential explanation why topoisomerase I cleavage lags behind that of classical caspase substrates such as poly(ADP-ribose) polymerase and lamin B1.


Assuntos
Apoptose , Caspases/metabolismo , DNA Topoisomerases Tipo I/metabolismo , Animais , Caspase 3 , Caspase 6 , Domínio Catalítico , Humanos , Células Jurkat , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Proteínas Recombinantes/metabolismo , Spodoptera
14.
J Clin Invest ; 103(1): 137-45, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9884343

RESUMO

Cholestatic liver injury appears to result from the induction of hepatocyte apoptosis by toxic bile salts such as glycochenodeoxycholate (GCDC). Previous studies from this laboratory indicate that cathepsin B is a downstream effector protease during the hepatocyte apoptotic process. Because caspases can initiate apoptosis, the present studies were undertaken to determine the role of caspases in cathepsin B activation. Immunoblotting of GCDC-treated McNtcp.24 hepatoma cells demonstrated cleavage of poly(ADP-ribose) polymerase and lamin B1 to fragments that indicate activation of effector caspases. Transfection with CrmA, an inhibitor of caspase 8, prevented GCDC-induced cathepsin B activation and apoptosis. Consistent with these results, an increase in caspase 8-like activity was observed in GCDC-treated cells. Examination of the mechanism of GCDC-induced caspase 8 activation revealed that dominant-negative FADD inhibited apoptosis and that hepatocytes isolated from Fas-deficient lymphoproliferative mice were resistant to GCDC-induced apoptosis. After GCDC treatment, immunoprecipitation experiments demonstrated Fas oligomerization, and confocal microscopy demonstrated DeltaFADD-GFP (Fas-associated death domain-green fluorescent protein, aggregation in the absence of detectable Fas ligand mRNA. Collectively, these data suggest that GCDC-induced hepatocyte apoptosis involves ligand-independent oligomerization of Fas, recruitment of FADD, activation of caspase 8, and subsequent activation of effector proteases, including downstream caspases and cathepsin B.


Assuntos
Apoptose/efeitos dos fármacos , Ácidos e Sais Biliares/farmacologia , Caspases/metabolismo , Catepsina B/metabolismo , Lamina Tipo B , Fígado/efeitos dos fármacos , Proteínas Virais , Receptor fas/metabolismo , Animais , Linhagem Celular , Inibidores de Cisteína Proteinase/farmacologia , Fragmentação do DNA/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Ácido Glicoquenodesoxicólico/farmacologia , Cinética , Laminas , Fígado/metabolismo , Camundongos , Proteínas Nucleares/metabolismo , Oligopeptídeos/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , RNA Mensageiro/genética , Ratos , Receptores do Fator de Necrose Tumoral/genética , Serpinas/genética
16.
Cancer Chemother Pharmacol ; 42(5): 391-9, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9771954

RESUMO

PURPOSE: To determine the effect of combined treatment with 7-ethyl-10-hydroxycamptothecin (SN-38, the active metabolite of irinotecan) and 5-fluorouracil/ folinic acid (5FU/FA) in vitro using HCT-8 human intestinal adenocarcinoma cells. METHODS: Cell survival was examined using colony forming assays. Cell cycle distribution before and after treatment was assessed by flow microfluorimetry. Levels of thymidylate synthase (TS) and topoisomerase I (topo I) in untreated and treated cells were determined by immunoblotting. Changes in deoxynucleotide pools were examined by high-performance liquid chromatography. RESULTS: Clonogenic assays revealed that colony formation was decreased by 50% after a 24-h exposure to 8+/-2 nM SN-38 or 12+/-3 microM 5FU, the latter being assayed in the presence of 2 microM FA. When treatment with 5FU/FA was followed by SN-38, the cytotoxicity was similar to that observed with 5FU/FA alone. In contrast, when HCT-8 cells were exposed to both agents simultaneously or to SN-38 followed by 5FU/FA, the cytotoxicity was greater than that of SN-38 or 5FU/FA treatment alone. Investigation of the mechanistic basis for this sequence dependence revealed that SN-38 treatment was associated with a dose- and time-dependent decrease in conversion of [5-3H]-2'-deoxyuridine to [3H]-H2O and thymidylate in intact cells. Immunoblotting failed to reveal any decrease in TS protein that could account for the decreased activity. High-performance liquid chromatography revealed that SN-38 treatment was associated with increased levels of the deoxynucleotide dTTP and decreased levels of dUTP. Flow microfluorimetry revealed that a 24-h treatment wit 10 nM SN-38 resulted in accumulation of HCT-8 cells in late S and G2 phases of the cell cycle, with a further increase in the G2 fraction during the 24 h after SN-38 removal. CONCLUSIONS: These observations are consistent with a model in which SN-38 sequentially induces diminished DNA synthesis, elevated dTTP pools, inhibition of dUMP synthesis and enhanced toxicity of 5FU/FA. Accordingly, sequencing of irinotecan and 5FU/FA might be important in combining these agents into an effective treatment for colorectal cancer.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Inibidores Enzimáticos/farmacologia , Inibidores da Topoisomerase I , Adenocarcinoma , Camptotecina/análogos & derivados , Camptotecina/farmacologia , Ciclo Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , DNA Topoisomerases Tipo I/metabolismo , Nucleotídeos de Desoxiuracil/metabolismo , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Fluoruracila/farmacologia , Humanos , Neoplasias Intestinais , Irinotecano , Leucovorina/farmacologia , Timidilato Sintase/antagonistas & inibidores , Nucleotídeos de Timina/metabolismo , Células Tumorais Cultivadas/efeitos dos fármacos
17.
Clin Cancer Res ; 4(3): 683-91, 1998 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9533538

RESUMO

Pyrazoloacridine (PA), an acridine congener with an unknown mechanism of action, has shown selective activity against solid tumor cells, cytotoxicity in noncycling and hypoxic cells, and promising antitumor activity in Phase I clinical trials. In the present study, the effect of PA on topoisomerase (topo) activity was evaluated using yeast strains lacking functional topo I or II, mammalian cell nuclear extracts, purified samples of mammalian topo I and topo II, and intact mammalian tissue culture cells. Clonogenic assays revealed that PA cytotoxicity in yeast strains was unaffected by selective loss of topo I or topo II activity. On the other hand, enzyme assays revealed that 2-4 microM PA abolished the catalytic activity of both topo I and topo II in vitro. In contrast to topotecan and etoposide, PA did not stabilize covalent topo-DNA complexes. Instead, PA inhibited topotecan-induced stabilization of covalent topo I-DNA complexes and etoposide-induced stabilization of topo II-DNA complexes in vitro and in intact cells. Consistent with these results, colony-forming assays indicated that short-term PA exposure inhibited the cytotoxicity of topotecan and etoposide, whereas prolonged PA exposure was itself toxic to these cells. Accumulation studies revealed that PA was concentrated as much as 250-fold in drug-treated cells, resulting in intranuclear concentrations that far exceeded those required to inhibit topo I and topo II. Collectively, these results not only suggest that PA can target both topo I and topo II at clinically achievable concentrations but also indicate that its mechanism is distinct from topo I and topo II poisons presently licensed for clinical use.


Assuntos
Acridinas/farmacologia , Acridinas/farmacocinética , Antineoplásicos/farmacologia , Antineoplásicos/farmacocinética , Pirazóis/farmacologia , Pirazóis/farmacocinética , Inibidores da Topoisomerase I , Inibidores da Topoisomerase II , Acridinas/toxicidade , Antineoplásicos/toxicidade , Sobrevivência Celular/efeitos dos fármacos , Doxorrubicina/toxicidade , Etoposídeo/toxicidade , Humanos , Leucemia Mieloide , Pirazóis/toxicidade , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/crescimento & desenvolvimento , Frações Subcelulares/metabolismo , Células Tumorais Cultivadas
18.
Clin Cancer Res ; 4(1): 117-30, 1998 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9516960

RESUMO

The nicotinamide analogue 6-aminonicotinamide (6AN) is presently undergoing evaluation as a potential modulator of the action of various antineoplastic treatments. Most previous studies of this agent have focused on a three-drug regimen of chemical modulators that includes 6AN. In the present study, the effect of single-agent 6AN on the efficacy of selected antineoplastic drugs was assessed in vitro. Colony-forming assays using human tumor cell lines demonstrated that pretreatment with 30-250 microM 6AN for 18 h resulted in increased sensitivity to the DNA cross-linking agent cisplatin, with 6-, 11-, and 17-fold decreases in the cisplatin dose that diminishes colony formation by 90% being observed in K562 leukemia cells, A549 non-small cell lung cancer cells, and T98G glioblastoma cells, respectively. Morphological examination revealed increased numbers of apoptotic cells after treatment with 6AN and cisplatin compared to cisplatin alone. 6AN also sensitized cells to melphalan and nitrogen mustard but not to chlorambucil, 4-hydroperoxycyclophosphamide, etoposide, or daunorubicin. In additional studies undertaken to elucidate the mechanism underlying the sensitization to cisplatin, atomic absorption spectroscopy revealed that 6AN had no effect on the rate of removal of platinum (Pt) adducts from DNA. Instead, 6AN treatment was accompanied by an increase in Pt-DNA adducts that paralleled the degree of sensitization. This effect was not attributable to 6AN-induced decreases in glutathione or NAD+, because other agents that depleted these detoxification cofactors (buthionine sulfoximine and 3-acetylpyridine, respectively) did not increase Pt-DNA adducts. On the contrary, 6AN treatment increased cellular accumulation of cisplatin. Further experiments revealed that 6AN was metabolized to 6-aminonicotinamide adenine dinucleotide (6ANAD+). Concurrent administration of nicotinamide and 6AN had minimal effect on cellular 6AN accumulation but abolished the formation of 6ANAD+, the increase in Pt-DNA adducts, and the sensitizing effect of 6AN in clonogenic assays. These observations identify 6AN as a potential modulator of cisplatin sensitivity and suggest that the 6AN metabolite 6ANAD+ exerts this effect by increasing cisplatin accumulation and subsequent formation of Pt-DNA adducts.


Assuntos
6-Aminonicotinamida/farmacologia , Antineoplásicos/farmacologia , Cisplatino/farmacologia , 6-Aminonicotinamida/metabolismo , Trifosfato de Adenosina/metabolismo , Adutos de DNA/metabolismo , Reparo do DNA/efeitos dos fármacos , Sinergismo Farmacológico , Humanos , NAD/metabolismo , Inibidores de Poli(ADP-Ribose) Polimerases , Células Tumorais Cultivadas
19.
Blood ; 91(3): 991-1000, 1998 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-9446661

RESUMO

Bcl-2, Bcl-xL, and Mcl-1 are three related intracellular polypeptides that have been implicated as negative regulators of apoptosis. In contrast, the partner protein Bax acts as a positive regulator of apoptosis. Based on the observation that all four of these polypeptides are expressed in a variety of acute myelogenous leukemia (AML) and acute lymphocytic leukemia (ALL) cell lines, cellular levels of these polypeptides were examined by immunoblotting in bone marrow samples harvested from 123 adult AML patients and 36 adult ALL patients before initial antileukemic therapy. Levels of Bcl-2, Mcl-1, Bcl-xL, and Bax each varied over a more than 10-fold range in different pretreatment leukemia specimens. When the 54 AML and 23 ALL samples that contained greater than 80% malignant cells were examined in greater detail, it was observed that pretreatment levels of Bcl-2 and Mcl-1 correlated with each other (R = .44, P < .001 for AML and R = .79, P < .0001 for ALL). In addition, a weak negative correlation between Bax expression and age was observed in AML samples (R = -0.35, P < .02) but not ALL samples. There was no relationship between pretreatment levels of these polypeptides and response to initial therapy. However, examination of 19 paired samples (the first harvested before chemotherapy and the second harvested 23 to 290 days later at the time of leukemic recurrence) revealed a greater than or equal to twofold increase in Mcl-1 levels in 10 of 19 pairs (7 of 15 AML and 3 of 4 ALL) at recurrence. In contrast, 2 of 19 pairs contained twofold less Mcl-1 at the time of recurrence. Approximately equal numbers of samples showed twofold increases and decreases in Bcl-2 (5 increases, 3 decreases) and Bcl-xL (1 increase, 4 decreases) at recurrence. Bax levels did not show a twofold decrease in any patient. these results, coupled with recent observations that cells overexpressing Mcl-1 are resistant to a variety of chemotherapeutic agents, raise the possibility that some chemotherapeutic regimens might select for leukemia cells with elevated levels of this particular apoptosis inhibitor.


Assuntos
Apoptose , Leucemia Mieloide Aguda/metabolismo , Proteínas de Neoplasias/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Adulto , Antineoplásicos/uso terapêutico , Medula Óssea/química , Resistencia a Medicamentos Antineoplásicos , Humanos , Immunoblotting , Leucemia Mieloide Aguda/tratamento farmacológico , Proteína de Sequência 1 de Leucemia de Células Mieloides , Proteínas de Neoplasias/análise , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Proteínas Proto-Oncogênicas/análise , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/análise , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Recidiva , Células Tumorais Cultivadas , Proteína X Associada a bcl-2
20.
Blood ; 90(11): 4283-96, 1997 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-9373239

RESUMO

Previous studies have shown that K562 chronic myelogenous leukemia cells are resistant to induction of apoptosis by a variety of agents, including the topoisomerase II (topo II) poison etoposide, when examined 4 to 24 hours after treatment with an initiating stimulus. In the present study, the responses of K562 cells and apoptosis-proficient HL-60 acute myelomonocytic leukemia cells to etoposide were compared, with particular emphasis on determining the long-term fate of the cells. When cells were treated with varying concentrations of etoposide for 1 hour and subsequently plated in soft agar, the two cell lines displayed similar sensitivities, with a 90% reduction in colony formation at 5 to 10 mu mol/L etoposide. After treatment with 17 mu mol/L etoposide for 1 hour, cleavage of the caspase substrate poly(ADP-ribose) polymerase (PARP), DNA fragmentation, and apoptotic morphological changes were evident in HL-60 cells in less than 6 hours. After the same treatment, K562 cells arrested in G2 phase of the cell cycle but otherwise appeared normal for 3 to 4 days before developing similar apoptotic changes. When the etoposide dose was increased to 68 mu mol/L, apoptotic changes were evident in HL-60 cells after 2 to 3 hours, whereas the same changes were observed in K562 cells after 24 to 48 hours. This delay in the development of apoptotic changes in K562 cells was accompanied by delayed release of cytochrome c to the cytosol and delayed appearance of peptidase activity that cleaved the fluorogenic substrates Asp-Glu-Val-Asp-aminotrifluoromethylcoumarin (DEVD-AFC) and Val-Glu-Ile-Asp-aminomethylcoumarin (VEID-AMC) as well as an altered spectrum of active caspases that were affinity labeled with N-(Nalpha-benzyloxycarbonylglutamyl-Nepsilon-biotin yllysyl) aspartic acid [(2,6-dimethylbenzoyl)oxy]methyl ketone [z-EK(bio)D-aomk]. On the other hand, the activation of caspase-3 under cell-free conditions occurred with indistinguishable kinetics in cytosol prepared from the two cell lines. Collectively, these results suggest that a delay in the signaling cascade upstream of cytochrome c release and caspase activation leads to a long latent period before the active phase of apoptosis is initiated in etoposide-treated K562 cells. Once the active phase of apoptosis is initiated, the spectrum and subcellular distribution of active caspase species differ between HL-60 and K562 cells, but a similar proportion of cells are ultimately killed in both cell lines.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Cisteína Endopeptidases/metabolismo , Etoposídeo/farmacologia , Eletroforese em Gel Bidimensional , Ativação Enzimática , Precursores Enzimáticos/metabolismo , Células HL-60 , Humanos , Cinética , Proteínas Proto-Oncogênicas c-abl/metabolismo , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...