Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
EBioMedicine ; 37: 205-213, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30385234

RESUMEN

BACKGROUND: Using patient-derived xenografts (PDXs) to assess chemosensitivity to anti-cancer agents in real-time may improve cancer care by enabling individualized clinical decision-making. However, it is unknown whether this new approach will be met with acceptance by patients, family and community. METHODS: We used a cross-sectional structured survey to investigate PDX acceptability with 1550 individuals across Australia and New Zealand (648 survivors of adult and childhood cancer, versus 650 community comparisons; and 48 parents of childhood cancer survivors versus 204 community parents). We identified factors influencing willingness-to-use PDXs, willingness-to-pay, maximum acceptable wait-time, and maximum acceptable number of mice used per patient. FINDINGS: PDXs were highly acceptable: >80% of those affected by cancer felt the potential advantages of PDXs outweighed the disadvantages (community participants: 68%). Survivors' and survivors' parents' most highly endorsed advantage was 'increased chance of survival'. 'Harm to animals' was the least endorsed disadvantage for all groups. Cancer survivors were more willing to use PDXs than community comparisons [p < ·001]. Survivors and survivors' parents were willing to pay more [p < ·001; p = ∙004 respectively], wait longer for results [p = ·03; p = ∙01], and use more mice [p = ·01; p < ∙001] than community comparisons. Male survivors found PDXs more acceptable [p = ·01] and were willing to pay more [p < ·001] than female survivors. Survivors with higher incomes found PDXs more acceptable [p = ·002] and were willing to pay more [p < ·001] than survivors with lower incomes. Mothers found PDXs more acceptable [p = ·04] but were less willing to wait [p = ·02] than fathers. INTERPRETATION: We found significant attitudinal support for PDX-guided cancer care. Willingness-to-pay and maximum acceptable number of mice align well with likely future usage. Maximum acceptable wait-times were lower than is currently achievable, highlighting an important area for future patient education until technology has caught up.


Asunto(s)
Supervivientes de Cáncer , Aceptación de la Atención de Salud , Medicina de Precisión/métodos , Ensayos Antitumor por Modelo de Xenoinjerto , Adulto , Animales , Femenino , Humanos , Masculino , Ratones , Proyectos Piloto , Factores Sexuales
2.
Mol Cell Biol ; 9(8): 3284-91, 1989 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-2552289

RESUMEN

Conditions, such as anoxia or glucose starvation, which induce the glucose-regulated set of stress proteins also lead to resistance to adriamycin (J. Shen, C. Hughes, C. Chao, J. Cai, C. Bartels, T. Gessner, and J. Subjeck, Proc. Natl. Acad. Sci. USA 84:3278-3282, 1987) and etoposide. We report here that chronic anoxia, glucose starvation, 2-deoxyglucose, the calcium ionophore A23187, glucosamine, ethylene glycol-bis(beta-aminoethyl ether)-N,N,N',N'-tetraacetic acid (EGTA), and tunicamycin (all specific inducers of the glucose regulated system) lead to a rapid and selective depletion of topoisomerase II from isolated nuclei of Chinese hamster ovary cells. This effect precedes a decline in tritiated thymidine incorporation and a redistribution of cells from S into G1/G0. The depletion of the enzyme is not accompanied by a decline in mRNA levels. We have also examined the mutant Chinese hamster K12 cell line which is temperature sensitive for expression of glucose-regulated proteins. When nuclei were isolated from K12 cells incubated at the nonpermissive temperature, a loss of topoisomerase II was again observed in congruence with the expression of stress proteins and cellular resistance to etoposide. These changes were not obtained in parental Wg1A cells incubated at the same temperature. These studies indicate that topoisomerase II is highly sensitive to glucose-regulated stresses and that its depletion from the nucleus, with the associated changes in cell cycle parameters, may represent general characteristics of the glucose-regulated state. Since anoxia and glucose starvation can occur during tumor development, this pathway for expression of drug resistance may have clinical ramifications.


Asunto(s)
ADN-Topoisomerasas de Tipo II/metabolismo , Regulación de la Expresión Génica , Glucosa/fisiología , Animales , Northern Blotting , Western Blotting , Ciclo Celular , Línea Celular , Núcleo Celular/metabolismo , Supervivencia Celular , Daño del ADN , Etopósido/farmacología , Proteínas de Choque Térmico/metabolismo , Calor , Oxígeno/fisiología , ARN Mensajero/metabolismo
3.
Cancer Res ; 52(7): 1817-22, 1992 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-1551112

RESUMEN

p34cdc2 kinase, an enzyme essential for mitosis in mammalian cells, may play a role in etoposide-induced G2 phase arrest of Chinese hamster ovary cells. In this study, etoposide is shown to cause inhibition of a specific p34cdc2 kinase activation pathway, that of tyrosine dephosphorylation. Exposure of asynchronously dividing cells to etoposide caused a simultaneous rapid decline of both mitotic index and p34cdc2 kinase activity, suggesting that the kinase was not activated and that the arrest point was in late G2 phase. Using synchronized cells, p34cdc2 kinase exhibited maximal activity at the G2/M transition. Activation of the kinase and the onset of mitosis were accompanied by increased electrophoretic mobility and tyrosine dephosphorylation of the p34cdc2 protein. A 1-h exposure to etoposide during early G2 phase inhibited p34cdc2 kinase activation, its shift in electrophoretic mobility, and its tyrosine dephosphorylation, all of which correlated with a delay in mitotic progression. The interaction between the p34cdc2 and cyclin B proteins appeared unaffected under etoposide exposure conditions which resulted in greater than 70% inhibition of p34cdc2 kinase activity and almost complete cessation of transition into mitosis. These data suggest that mammalian cells express a DNA damage-responsive mechanism which controls mitotic progression at the level of p34cdc2 tyrosine dephosphorylation.


Asunto(s)
Proteína Quinasa CDC2/metabolismo , Etopósido/farmacología , Índice Mitótico/efectos de los fármacos , Animales , Western Blotting , Proteína Quinasa CDC2/antagonistas & inhibidores , Células CHO , Ciclo Celular/efectos de los fármacos , Cricetinae , Electroforesis en Gel de Poliacrilamida , Activación Enzimática , Fase G2/efectos de los fármacos , Cinética , Metionina/metabolismo , Biosíntesis de Proteínas , Proteínas/aislamiento & purificación , Factores de Tiempo
4.
Cancer Res ; 56(17): 4006-12, 1996 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-8752171

RESUMEN

The Bcl-2 oncoprotein, which is expressed in a variety of human malignancies, blocks apoptosis induced by chemotherapeutic drugs, including the topoisomerase II inhibitor, etoposide. To determine the significance of Bcl-2 in etoposide-induced death of human epithelial tumor cells, HeLa S3 cells were transfected with human bcl-2 cDNA in the pSFFV expression vector, and stable Bcl-2-expressing clones established. In agreement with previous studies, Bcl-2 inhibited loss of cell viability (by trypan blue exclusion), the appearance of morphologically apoptotic cells, and the amount of low molecular weight DNA extracted after etoposide exposure (25 microns, 4 h). The degree of inhibition, compared to wild-type and vector control-transfected clones, differed according to the level of Bcl-2 protein expressed in the two clones studied. However, when cell survival was assessed by colony-forming assays, no significant differences were detected at any of the etoposide concentrations used. Although Bcl-2 inhibited etoposide-induced apoptosis, it had no effect on the formation of giant, multinucleated cells characteristic of mitotic catastrophe. Consequently, the ability of Bcl-2 to prevent apoptosis caused by chemotherapeutic drugs may not necessarily translate into increased survival of cells that express Bcl-2.


Asunto(s)
Apoptosis/efectos de los fármacos , Etopósido/farmacología , Neoplasias/tratamiento farmacológico , Proteínas Proto-Oncogénicas/fisiología , Apoptosis/fisiología , Muerte Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Epitelio/efectos de los fármacos , Epitelio/patología , Células HeLa , Humanos , Neoplasias/patología , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-bcl-2 , Transfección
5.
Cancer Res ; 50(12): 3761-6, 1990 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-2187601

RESUMEN

The mammalian homologue of the yeast cdc2 gene product, p34cdc2, is a cell cycle-regulated protein essential for mitosis. We have used polyclonal antisera raised against a peptide corresponding to the carboxyl terminus of the sequence of human cdc2 to study p34cdc2 in Chinese hamster ovary (CHO) cells. Major bands are immunoprecipitated at a molecular weight of 34,000, although not in the presence of competing antigenic peptide. p34cdc2 is coimmunoprecipitated with proteins of molecular weights of 52,000 and 57,000. Immunoprecipitates express histone H1 kinase activity which varies throughout the cell cycle, maximal activity being observed in G2-M. The activity of the p34cdc2 kinase varies according to its association with the Mr 52,000 and 57,000 proteins and according to their phosphorylation state. Treatment of either asynchronous CHO cells or an enriched G2 population with the antitumor agent, etoposide, results in rapid inhibition of immunoprecipitated p34cdc2 kinase activity, which is not due to a direct effect of drug upon the enzyme. p34cdc2 kinase activity recovers as cells arrest in G2 and a second etoposide treatment further inhibits p34cdc2 kinase activity and prolongs G2 arrest. Exposure of asynchronous CHO cells to gamma-irradiation also inhibits p34cdc2 kinase activity within 1 h. Again this activity recovers as cells accumulate in G2. These results suggest that DNA damage in CHO cells elicits a response which results in inhibition of p34cdc2 kinase activity and, consequently, G2 arrest.


Asunto(s)
Ciclo Celular , Fosfoproteínas/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas , Animales , Proteína Quinasa CDC2 , Ciclo Celular/efectos de los fármacos , Ciclo Celular/efectos de la radiación , Línea Celular , Cricetinae , ADN/efectos de la radiación , Daño del ADN , Etopósido/farmacología , Peso Molecular , Fosfoproteínas/análisis , Fosfoproteínas/metabolismo , Fosfoproteínas/efectos de la radiación , Fosforilación , Proteínas Quinasas/análisis , Proteínas Quinasas/metabolismo , Proteínas Quinasas/efectos de la radiación , Factores de Tiempo
6.
Cancer Res ; 50(12): 3767-71, 1990 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-2187602

RESUMEN

In an effort to shed light upon the processes of antitumor drug-induced cell death, we have carried out a systemic study of the effects of the anti-topoisomerase II agent, etoposide, on Chinese hamster ovary cells. Treatment of Chinese hamster ovary cells for 1 h with a 2-log cell-killing concentration of etoposide induces a high incidence of DNA single-strand breaks which are rapidly repaired upon drug removal. p34cdc2 kinase activity is inhibited within 1 h of addition of etoposide. Following removal of drug, cells accumulate transiently in G2. Upon recovery of p34cdc2 kinase activity (between 12 and 24 h posttreatment), approximately 50% of cells progress through mitosis which results in micronucleation. Examination of mitotic figures at various posttreatment incubation times indicates that micronucleation of daughter cells could be attributed to abnormal segregation of chromosomes during mitosis. Unexpectedly, p34cdc2 kinase activity remains elevated relative to untreated controls until 36 h post-etoposide treatment, a point where no further cell division takes place. This activity decreases by 48 h posttreatment, concomitant with a decrease in cell viability as estimated by the ability to exclude trypan blue. These results indicate that etoposide may induce cytotoxicity via gross chromosomal fragmentation, and that p34cdc2 kinase may be involved in this process.


Asunto(s)
Supervivencia Celular , Fosfoproteínas/aislamiento & purificación , Proteínas Quinasas/aislamiento & purificación , Animales , Proteína Quinasa CDC2 , Ciclo Celular/efectos de los fármacos , Ciclo Celular/efectos de la radiación , Línea Celular , Cricetinae , Etopósido/farmacología , Peso Molecular , Fosfoproteínas/antagonistas & inhibidores , Fosfoproteínas/efectos de la radiación , Inhibidores de Proteínas Quinasas , Proteínas Quinasas/efectos de la radiación
7.
Cancer Res ; 54(18): 4933-9, 1994 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-8069859

RESUMEN

Morphological examination of HeLa cells exposed to etoposide for 1 h revealed two distinct modes of death: (a) within 6 h of drug removal, shrunken cells appeared which contained vacuolated cytoplasm and regions of intense chromatin staining, consistent with apoptosis; and (b) concomitant with release from G2 arrest, enlarged cells appeared which contained evenly staining nuclear fragments, consistent with mitotic death. The methylxanthine, caffeine, enhanced cytotoxicity in a concentration-dependent manner when applied for 24 h following etoposide exposure. One mM caffeine alleviated etoposide-induced G2 arrest and increased the incidence of mitotic death, accounting for the potentiation of cytotoxicity. Brief caffeine exposures (5 or 10 mM for 1-2 h) caused specific tyrosine dephosphorylation and activation of p34cdc2 kinase, and mitotic progression to a limited extent, in cells which were arrested in G2 following etoposide treatment. However, longer exposure times at a high caffeine concentration (10 mM) caused inhibition of both cell cycle progression and mitotic death, and the enhancement of etoposide cytotoxicity could be accounted for by up to a 3-fold increase in the proportion of morphologically apoptotic cells. Thus, caffeine potentiates etoposide cytotoxicity by two morphologically distinct mechanisms depending on its concentration.


Asunto(s)
Cafeína/farmacología , Etopósido/farmacología , Fase G2/efectos de los fármacos , Células HeLa/efectos de los fármacos , Apoptosis/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Células HeLa/patología , Humanos , Fosforilación , Factores de Tiempo , Tirosina/metabolismo
8.
Cancer Res ; 51(24): 6543-9, 1991 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-1660341

RESUMEN

A new multiple drug-resistant Chinese hamster ovary cell line, CHO-SMR5, has been isolated which demonstrates a direct correlation between reduced cellular topoisomerase II activity (5-fold reduction) and a low level of resistance (3- to 7-fold) to topoisomerase II inhibitors. This cell line, initially selected for resistance to 9-(4,6-O-ethylidene-beta-D-glucopyranosyl)-4'-demethylepipodophylloto xin, exhibits cross-resistance to other topoisomerase II inhibitors including 4'-(9-acridinylamino)methanesulfon-m-anisidide, doxorubicin, and mitoxantrone. The resistant cells show a 4.5-fold decrease in topoisomerase II by immunoblotting when compared to wild-type cells. Drug uptake studies reveal equivalent equilibrium intracellular concentrations of [3H]9-(4,6-O-ethylidene-beta-D-glucopyranosyl)-4'-demethyepipodophyll otoxin in the resistant and parental cells. The catalytic activity of topoisomerase II (decatenation of kinetoplast DNA) is 5-fold less in the drug-resistant cell line relative to wild-type Chinese hamster ovary cells. Drug-induced DNA damage, measured as either formation of DNA double-strand breaks or covalent DNA-enzyme complexes, is 4-fold less in the resistant cell line. Finally, Northern blot analysis demonstrates a 5-fold reduction in topoisomerase II mRNA isolated from log phase CHO-SMR5 cells. These findings suggest that a reduced level of topoisomerase II is likely to be the sole mechanism of drug resistance in this novel cell line.


Asunto(s)
ADN-Topoisomerasas de Tipo II/metabolismo , Etopósido/farmacología , Animales , Transporte Biológico , Northern Blotting , Western Blotting , Células CHO , Núcleo Celular/enzimología , Cricetinae , Daño del ADN , ADN-Topoisomerasas de Tipo II/genética , ADN-Topoisomerasas de Tipo II/inmunología , ADN Superhelicoidal/metabolismo , Resistencia a Medicamentos , Expresión Génica , ARN Mensajero/genética , Inhibidores de Topoisomerasa II
9.
Cancer Res ; 48(22): 6404-10, 1988 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-2846151

RESUMEN

In Chinese hamster ovary cells, stable mutants that exhibit 250- to 350-fold resistance to camptothecin (CptR mutants) have been isolated from mutagen-treated cultures. The CptR mutants exhibited no cross-resistance towards drugs such as colchicine, vinblastine, taxol, or puromycin but showed slightly (2- to 3-fold) enhanced sensitivity towards various drugs that inhibit DNA topoisomerase II (namely teniposide, etoposide, doxorubicin, mitoxantrone, amsacrine, ellipticine), suggesting that the genetic lesion in these mutants was highly specific. In contrast to the wild-type cells, the CptR line was resistant to camptothecin-induced DNA strand breaks as measured by alkaline elution. Biochemical studies revealed that in CptR mutants the cellular activity as well as protein content of DNA topoisomerase I were reduced to about 40-50% of the level in wild-type cells. Normal levels of activity and content were observed for the related enzyme DNA topoisomerase II. Studies with DNA topoisomerase I purified from the wild-type and the mutant cells showed that the enzyme from the CptR cells was markedly resistant to camptothecin as assayed by the drug's effects either on relaxation of supercoiled DNA or on stabilization of the covalent enzyme-DNA intermediate. The presence of a camptothecin-resistant form of DNA topoisomerase I in the mutant cells provides further evidence that this enzyme is the cellular target of camptothecin. Cell hybridization studies between the CptR and CptS cells showed that the hybrids formed between these two cell lines were sensitive to camptothecin. The recessive behavior of the CptR mutation provides a plausible explanation for the reduced topoisomerase I content (about one-half of wild-type cells) of the mutant cells and also for their enhanced sensitivity towards inhibitors of topoisomerase II.


Asunto(s)
Camptotecina/farmacología , ADN-Topoisomerasas de Tipo I/análisis , Animales , Cricetinae , Cricetulus , Daño del ADN , ADN-Topoisomerasas de Tipo I/aislamiento & purificación , ADN-Topoisomerasas de Tipo II/análisis , Resistencia a Medicamentos , Células Híbridas , Mutación
10.
Leukemia ; 30(11): 2133-2141, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27416986

RESUMEN

Acute lymphoblastic leukemia (ALL) in children exemplifies how multi-agent chemotherapy has improved the outcome for patients. Refinements in treatment protocols and improvements in supportive care for this most common pediatric malignancy have led to a cure rate that now approaches 90%. However, certain pediatric ALL subgroups remain relatively intractable to treatment and many patients who relapse face a similarly dismal outcome. Moreover, survivors of pediatric ALL suffer the long-term sequelae of their intensive treatment throughout their lives. Therefore, the development of drugs to treat relapsed/refractory pediatric ALL, as well as those that more specifically target leukemia cells, remains a high priority. As pediatric malignancies represent a minority of the overall cancer burden, it is not surprising that they are generally underrepresented in drug development efforts. The identification of novel therapies relies largely on the reappropriation of drugs developed for adult malignancies. However, despite the large number of experimental agents available, clinical evaluation of novel drugs for pediatric ALL is hindered by limited patient numbers and the availability of effective established drugs. The Pediatric Preclinical Testing Program (PPTP) was established in 2005 to provide a mechanism by which novel therapeutics could be evaluated against xenograft and cell line models of the most common childhood malignancies, including ALL, to prioritize those with the greatest activity for clinical evaluation. In this article, we review the results of >50 novel agents and combinations tested against the PPTP ALL xenografts, highlighting comparisons between PPTP results and clinical data where possible.


Asunto(s)
Antineoplásicos/uso terapéutico , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Niño , Humanos , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Terapia Recuperativa/tendencias , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Leukemia ; 30(7): 1552-61, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27055868

RESUMEN

Aberrant ERG (v-ets avian erythroblastosis virus E26 oncogene homolog) expression drives leukemic transformation in mice and high expression is associated with poor patient outcomes in acute myeloid leukemia (AML) and T-acute lymphoblastic leukemia (T-ALL). Protein phosphorylation regulates the activity of many ETS factors but little is known about ERG in leukemic cells. To characterize ERG phosphorylation in leukemic cells, we applied liquid chromatography coupled tandem mass spectrometry and identified five phosphorylated serines on endogenous ERG in T-ALL and AML cells. S283 was distinct as it was abundantly phosphorylated in leukemic cells but not in healthy hematopoietic stem and progenitor cells (HSPCs). Overexpression of a phosphoactive mutant (S283D) increased expansion and clonogenicity of primary HSPCs over and above wild-type ERG. Using a custom antibody, we screened a panel of primary leukemic xenografts and showed that ERG S283 phosphorylation was mediated by mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) signaling and in turn regulated expression of components of this pathway. S283 phosphorylation facilitates ERG enrichment and transactivation at the ERG +85 HSPC enhancer that is active in AML and T-ALL with poor prognosis. Taken together, we have identified a specific post-translational modification in leukemic cells that promotes progenitor proliferation and is a potential target to modulate ERG-driven transcriptional programs in leukemia.


Asunto(s)
Leucemia/patología , Sistema de Señalización de MAP Quinasas/fisiología , Sitios de Unión , Línea Celular Tumoral , Proliferación Celular , Células Madre Hematopoyéticas , Humanos , Leucemia Mieloide Aguda/patología , Fosforilación , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Procesamiento Proteico-Postraduccional , Serina/metabolismo , Regulador Transcripcional ERG/metabolismo , Transcriptoma
12.
Oncogene ; 18(44): 5991-9, 1999 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-10557088

RESUMEN

Ligation of the Fas cell surface receptor leads to activation of caspases and subsequent apoptosis. Members of the Bcl-2 family of proteins control the cellular commitment to apoptosis, although their role in Fas-induced apoptosis is ill-defined. In this report we demonstrate that the pro-apoptotic protein, Bax, translocates from the cytosol specifically to the mitochondria following Fas ligation in MCF10A1 breast epithelial cells. Bax translocation was dependent on caspase activation, and preceded the release of cytochrome c and loss of mitochondrial respiratory activity. Bax translocation occurred in concert with activation of downstream caspases as determined by cleavage of a synthetic substrate, proteolysis of poly(ADP-ribose) polymerase, and processing of procaspase-3 and -7. Overexpression of the anti-apoptotic protein, Bcl-2, prevented Bax insertion, cytochrome c release, complete processing of caspase-3 and -7, and full activation of DEVD-specific cleavage activity. These data establish a role for Bax mitochondrial insertion during Fas-mediated apoptosis, and support a model in which Bax insertion amplifies the Fas apoptotic cascade through cytochrome c release and complete processing of caspases-3 and -7. In addition, our findings indicate that prevention of Bax insertion into the mitochondria represents a novel mechanism by which Bcl-2 inhibits Fas-induced apoptosis.


Asunto(s)
Apoptosis/fisiología , Grupo Citocromo c/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptor fas/metabolismo , Clorometilcetonas de Aminoácidos/farmacología , Transporte Biológico , Mama/citología , Caspasa 3 , Caspasa 7 , Inhibidores de Caspasas , Caspasas/metabolismo , Respiración de la Célula , Inhibidores de Cisteína Proteinasa/farmacología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Regulación de la Expresión Génica , Humanos , Membranas Intracelulares/metabolismo , Mitocondrias/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteína X Asociada a bcl-2
13.
Cell Death Differ ; 7(1): 102-11, 2000 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10713725

RESUMEN

The pro-apoptotic protein, Bax, has been reported to translocate from cytosol to mitochondria following exposure of cells to apoptotic stresses including cytokine withdrawal and treatment with glucocorticoids and cytotoxic drugs. These observations, coupled with reports showing that Bax causes the release of mitochondrial cytochrome c, implicate Bax as a central mediator of the apoptotic process. In this report we demonstrate by subcellular fractionation a significant shift in Bax localization from cytosol to cellular membranes in two human tumor cell lines exposed to staurosporine or etoposide. Immunofluorescence studies confirmed that Bax specifically relocalized to the mitochondria. This redistribution of Bax occurred in concert with, or just prior to, proteolytic processing of procaspase-3, activation of DEVD-specific cleavage activity and degradation of poly(ADP-ribose) polymerase. However, Bax membrane translocation was independent of caspase activity as determined using the broad-range caspase inhibitor z-VAD-fmk. High level overexpression of the anti-apoptotic protein Bcl-2 prevented Bax redistribution to the mitochondria, caspase activation and apoptosis following exposure to staurosporine or etoposide. These data confirm the role of Bax in mitochondrial cytochrome c release, and indicate that prevention of Bax translocation to the mitochondrial membrane represents a novel mechanism by which Bcl-2 inhibits drug-induced apoptosis.


Asunto(s)
Apoptosis , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Transporte Biológico , Caspasa 3 , Inhibidores de Caspasas , Grupo Citocromo c/metabolismo , Citosol/metabolismo , Etopósido/farmacología , Expresión Génica , Células HL-60 , Células HeLa , Humanos , Membranas Intracelulares/metabolismo , Mitocondrias/metabolismo , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Inhibidores de Proteínas Quinasas , Proteínas Proto-Oncogénicas c-bcl-2/genética , Estaurosporina/farmacología , Células Tumorales Cultivadas , Proteína X Asociada a bcl-2
14.
Leukemia ; 15(6): 929-35, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11417479

RESUMEN

The molecular basis for the clinical presentation of broad-range drug resistance in childhood ALL is poorly understood. In this study, high level cross-resistance to the glucocorticoid dexamethasone was encountered in a childhood ALL cell line selected for resistance to methotrexate (CEM MTX-R3). Compared with wild-type (WT) CEM cells, MTX-R3 cells had significantly fewer glucocorticoid binding sites, as well as reduced glucocorticoid receptor protein and mRNA levels. DNA sequencing and restriction fragment-length polymorphism (RFLP) analysis showed that WT cells expressed both a wild-type and a mutant (GR753F) glucocorticoid receptor allele, while MTX-R3 cells expressed only the GR753F allele. Therefore, the cross-resistance of MTX-R3 cells to dexamethasone appeared due to loss of expression of the wild-type glucocorticoid receptor allele. In an effort to gain insight into the underlying basis for the development of cross-resistance to methotrexate and glucocorticoids, glucocorticoid receptor nuclear translocation experiments were carried out. Exposure of WT cells to either dexamethasone or the cytotoxic agents cytarabine and methotrexate caused translocation of the glucocorticoid receptor from the cytoplasm into the nucleus. These data indicate that exposure of childhood ALL cells to cytotoxic agents may result in ligand-independent glucocorticoid receptor activation which, in the context of the outgrowth of drug-resistant cells, could lead to the co-selection of glucocorticoid resistance.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Múltiples Medicamentos/genética , Resistencia a Antineoplásicos/genética , Glucocorticoides/farmacología , Metotrexato/farmacología , Proteínas de Neoplasias/deficiencia , Células Madre Neoplásicas/efectos de los fármacos , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Receptores de Glucocorticoides/deficiencia , Transporte Activo de Núcleo Celular/efectos de los fármacos , Alelos , Niño , Citarabina/farmacología , Análisis Mutacional de ADN , ADN de Neoplasias/genética , Dexametasona/farmacología , Humanos , Leucemia de Células T/genética , Leucemia de Células T/patología , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Polimorfismo de Longitud del Fragmento de Restricción , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Unión Proteica , ARN Mensajero/genética , ARN Neoplásico/genética , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Selección Genética , Análisis de Secuencia de ADN , Células Tumorales Cultivadas/efectos de los fármacos
15.
Leukemia ; 29(4): 819-27, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25306899

RESUMEN

High expression of the ETS family transcription factor ERG is associated with poor clinical outcome in acute myeloid leukemia (AML) and acute T-cell lymphoblastic leukemia (T-ALL). In murine models, high ERG expression induces both T-ALL and AML. However, no study to date has defined the effect of high ERG expression on primary human hematopoietic cells. In the present study, human CD34+ cells were transduced with retroviral vectors to elevate ERG gene expression to levels detected in high ERG AML. RNA sequencing was performed on purified populations of transduced cells to define the effects of high ERG on gene expression in human CD34+ cells. Integration of the genome-wide expression data with other data sets revealed that high ERG drives an expression signature that shares features of normal hematopoietic stem cells, high ERG AMLs, early T-cell precursor-ALLs and leukemic stem cell signatures associated with poor clinical outcome. Functional assays linked this gene expression profile to enhanced progenitor cell expansion. These results support a model whereby a stem cell gene expression network driven by high ERG in human cells enhances the expansion of the progenitor pool, providing opportunity for the acquisition and propagation of mutations and the development of leukemia.


Asunto(s)
Transformación Celular Neoplásica/genética , Sangre Fetal/metabolismo , Células Madre/metabolismo , Transactivadores/genética , Transcriptoma , Antígenos CD34/genética , Antígenos CD34/metabolismo , Proliferación Celular , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Células Cultivadas , Sangre Fetal/citología , Regulación de la Expresión Génica , Vectores Genéticos , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Mutación , Retroviridae/genética , Retroviridae/metabolismo , Células Madre/citología , Transactivadores/metabolismo , Regulador Transcripcional ERG , Transducción Genética
16.
Eur J Cancer ; 37(17): 2217-21, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11677110

RESUMEN

The clinical aggressiveness of neuroblastoma, a childhood embryonal tumour of neuroectodermal cells derived from the neural crest, is considered to be dictated by the competitive interactions between cell proliferation, differentiation and apoptosis. Caspase-9 is a central effector enzyme in the apoptotic mechanism. Recent studies with caspase-9 (CASP9) knockout mice indicate a primary defect in the brain caused by decreased apoptosis during the early stages of nervous system development. It is our hypothesis that silencing of CASP9 through genetic mutations may promote neuroblastoma tumorigenesis. Here, we report the outcome of screening neuroblastoma tumours for silencing mutations in CASP9. cDNA prepared from RNA isolated from 22 neuroblastoma tumours representing the full range of neuroblastoma clinicopathological disease stages was sequenced. Single nucleotide changes were detected in all neuroblastoma tumours, but were found not to represent silencing mutations, but rather sequence polymorphisms. These polymorphisms did not associate with the clinicopathological stages of disease or the predicted clinical outcomes of the patients. Silencing mutations of CASP9 are therefore unlikely to be causal to neuroblastoma tumorigenesis.


Asunto(s)
Caspasas/genética , Genes Supresores de Tumor , Neuroblastoma/genética , Secuencia de Bases , Caspasa 9 , Transformación Celular Neoplásica/genética , Niño , ADN Complementario/genética , ADN de Neoplasias/genética , Silenciador del Gen , Humanos , Datos de Secuencia Molecular , Mutación , Polimorfismo de Nucleótido Simple
17.
Cancer Chemother Pharmacol ; 41(6): 457-63, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-9554589

RESUMEN

UNLABELLED: A reduced capacity for apoptosis induction is considered to play a significant role both in the development of malignancy and in tumor cell resistance to chemotherapeutic drugs. The Bcl-2 oncoprotein inhibits apoptosis induced by antitumor agents at a point downstream of drug-target interactions. Stable expression of Bcl-2 in the human epithelial tumor (HeLa) cell line results in inhibition of apoptosis following exposure to the topoisomerase II poison, etoposide. However, Bcl-2 is unable to enhance clonogenic survival as a result of alternate pathways to reproductive death induced by the drug. PURPOSE: The purpose of this study was to further investigate the role of Bcl-2 in human epithelial tumor cell drug resistance using 5-fluoro-2'-deoxyuridine, staurosporine, and doxorubicin, in addition to etoposide. METHODS: The ability of Bcl-2 to enhance clonogenic cell survival was studied by colony-forming assays, while delay of cell death induction was assessed by trypan blue viability measurements. The proportion of apoptotic cells was measured by morphological criteria, as well as detection of apoptotic DNA fragmentation using the terminal deoxynucleotidyl transferase assay. RESULTS: Despite profound inhibition to loss of plasma membrane integrity for all agents tested, Bcl-2 was only able to significantly increase clonogenic survival following exposure to 5-fluoro-2'-deoxyuridine and staurosporine, but not following exposure to etoposide or doxorubicin. Furthermore, the time-course of apoptosis induction following exposure of HeLa cells to equitoxic concentrations of staurosporine and etoposide was profoundly different. CONCLUSIONS: These results indicate that Bcl-2 enhances clonogenic survival of human epithelial tumor cells in an agent-specific fashion, which may be determined by the initial cytotoxic lesion induced by a particular drug.


Asunto(s)
Apoptosis/efectos de los fármacos , Doxorrubicina/toxicidad , Etopósido/toxicidad , Floxuridina/toxicidad , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Estaurosporina/toxicidad , Antineoplásicos/toxicidad , Supervivencia Celular/efectos de los fármacos , Fragmentación del ADN/efectos de los fármacos , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Células HeLa/efectos de los fármacos , Células HeLa/metabolismo , Humanos
18.
Cancer Chemother Pharmacol ; 39(5): 399-409, 1997.
Artículo en Inglés | MEDLINE | ID: mdl-9054953

RESUMEN

UNLABELLED: Protein kinase inhibitors have demonstrated potential for use in the therapy of human cancers, in particular leukemia. Staurosporine, a protein kinase inhibitor with broad specificity, enhances the cytotoxic effects of various antitumor agents with different modes of action. The topoisomerase II inhibitor, etoposide, has shown clinical activity against a wide range of tumor types. PURPOSE: The purpose of this study was to assess the effects of staurosporine on etoposide-induced cell death processes in a human tumor of epithelial origin. METHODS: Modulation of etoposide-induced apoptosis by staurosporine in HeLa cells was assessed by cell morphology, extraction of low molecular weight DNA, quantitation of DNA-protein complexes, and measurements of rates of DNA synthesis. The effects on cellular genes implicated in apoptosis were determined by Northern and Western blotting, along with assays of cyclin-dependent kinase activities. RESULTS: Staurosporine exhibited a two- to three-fold potentiation of apoptosis caused by etoposide in HeLa cells when applied concurrently, or immediately following etoposide removal, but did not alter the quantity of DNA-protein complexes produced by etoposide. Etoposide-induced apoptosis, and its potentiation by staurosporine, were associated with reduced c-myc expression, and a moderate increase in p21WAF1/CIP1 mRNA and protein levels. Inhibitors of cyclic AMP-dependent protein kinase and protein kinase C, which exhibit greater specificity than staurosporine, were without effect on apoptosis caused by etoposide, whereas use of the tyrosine phosphatase inhibitor, vanadate, resulted in its abrogation. The potentiation of etoposide-induced apoptosis by staurosprine was associated with a significant increase in cyclin A-dependent kinase activity. In addition, etoposide caused substantial inhibition of DNA synthesis. CONCLUSION: These results indicate that staurosporine potentiates apoptosis through events which occur downstream of DNA damage, and implicate unscheduled activation of cyclin A-dependent kinase during inhibition of DNA synthesis as a possible cause.


Asunto(s)
Apoptosis/efectos de los fármacos , Quinasas CDC2-CDC28 , Etopósido/toxicidad , Proteínas de Neoplasias/metabolismo , Estaurosporina/farmacología , Neoplasias de la Mama , Proteína Quinasa CDC2/metabolismo , Línea Celular , Quinasa 2 Dependiente de la Ciclina , Quinasas Ciclina-Dependientes/metabolismo , Replicación del ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Femenino , Células HeLa , Humanos , Cinética , Proteínas de Neoplasias/efectos de los fármacos , Proteínas de Neoplasias/aislamiento & purificación , Proteínas Serina-Treonina Quinasas/metabolismo , Células Tumorales Cultivadas
19.
Cancer Chemother Pharmacol ; 44(1): 1-11, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10367743

RESUMEN

UNLABELLED: Defective apoptotic mechanisms are considered to play a role in both the development of malignancy and resistance to chemotherapeutic drugs. The Bcl-2 family of proteins regulate the cellular commitment to survive or die when challenged with various apoptotic stimuli. PURPOSE: The purpose of this study was to identify the point at which Bcl-2 interrupts the apoptotic cascade initiated following exposure of human tumor cells to etoposide. METHODS: A stable Bcl-2-expressing HeLa-transfected clonal cell line, along with its control-vector-transfected counterpart, were utilized in this study. Following etoposide exposure, cells were examined for cell cycle arrest, formation of hyperdiploid cells, apoptotic DNA degradation, loss of plasma membrane integrity, levels of expression of members of the Bcl-2 protein family, caspase activation, degradation of poly(ADP-ribose) polymerase and movement of Bax from cytosol to cellular membrane fractions. RESULTS: Caspase activation, poly(ADP-ribose) polymerase degradation and Bax membrane insertion were initiated rapidly following etoposide removal, concomitantly with cell cycle arrest. Whereas Bcl-2 had no effect on etoposide-induced cell arrest, it interrupted all aspects of apoptosis, including activation of caspases, poly(ADP-ribose) polymerase degradation, DNA fragmentation and loss of plasma membrane integrity. Surprisingly, Bcl-2 also blocked Bax membrane insertion. In addition, Bcl-2 also prevented the increase in cellular levels of Bak, Bax and Bcl-xL, along with degradation of actin and Bax. However, inhibition of etoposide-induced apoptosis by Bcl-2 resulted in the accumulation of giant, multinucleated cells that eventually lost the ability to exclude trypan blue without apoptotic morphology or DNA degradation. CONCLUSIONS: These results indicate that biochemical apoptotic processes are initiated concomitant with etoposide-induced cell cycle arrest and are interrupted by Bcl-2 overexpression. However, the aberrant mitotic events induced by etoposide are sufficient to kill these cells even in the absence of apoptosis.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Transformación Celular Neoplásica , Etopósido/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Apoptosis/fisiología , Caspasas/efectos de los fármacos , Caspasas/metabolismo , Núcleo Celular/efectos de los fármacos , Núcleo Celular/ultraestructura , Genes bcl-2/genética , Células HeLa/efectos de los fármacos , Humanos , Mitosis/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/farmacología
20.
Cancer Chemother Pharmacol ; 44(4): 275-82, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10447574

RESUMEN

PURPOSE: Methyl N-(4'-(9-acridinylamino)-phenyl)carbamate hydrochloride (AMCA) and methyl N-(4'-(9-acridinylamino)-2-methoxyphenyl)carbamate hydrochloride (mAMCA) are analogues of the topoisomerase II (topo II) poison amsacrine, and are distinguished from amsacrine by their high cytotoxicity towards non-cycling cells. Since mammalian cells contain two forms (alpha and beta) of topo II and the alpha isoform is down-regulated in non-cycling cells, we have considered whether these carbamate analogues target topo IIbeta selectively. METHODS: A drug permeable yeast strain (JN394 top2-4) was transformed using a shuttle vector containing either human top2alpha, human top2alpha or yeast top2 under the control of a GAL1 promoter. The strain was analysed at a non-permissive temperature, where only the plasmid-borne topo II was active. RESULTS: AMCA and mAMCA produced comparable levels of cell killing with human DNA topo IIalpha, human DNA topo IIbeta and yeast DNA topo II. Two other acridine derivatives N-[2-(dimethylamino)ethyl]acridine-4-carboxamide (DACA) and its 7-chloro derivative, which like AMCA and mAMCA are able to overcome multidrug resistance mechanisms, were much more active against human DNA topo IIalpha than against human DNA topo IIbeta and yeast DNA topo II. A series of mutant Chinese hamster and human lines with defined topo lesions, including the HL60/MX2 line that lacks topo IIbeta expression, was also used to compare resistance to amsacrine, AMCA and etoposide. Loss of topo IIbeta activity had a greater effect on amsacrine and AMCA than on etoposide. Resistance of murine Lewis lung cultures in exponential and plateau phase was also measured. Loss of topo IIalpha activity, as measured in both mutant cells expressing lower amounts of enzyme and in cells in plateau phase, resulted in concomitant acquisition of resistance that was greatest for etoposide and least for AMCA. CONCLUSION: We conclude that the carbamate analogues of amsacrine recognize both topo IIalpha and beta in cells.


Asunto(s)
Amsacrina/análogos & derivados , Antineoplásicos/farmacología , ADN-Topoisomerasas de Tipo II , Isoenzimas/antagonistas & inhibidores , Inhibidores de Topoisomerasa II , Acridinas/farmacología , Amsacrina/farmacología , Animales , Antígenos de Neoplasias , Células CHO , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Carcinoma Pulmonar de Lewis/enzimología , Ciclo Celular/efectos de los fármacos , Cricetinae , ADN-Topoisomerasas de Tipo II/aislamiento & purificación , Proteínas de Unión al ADN , Etopósido/farmacología , Humanos , Isoenzimas/aislamiento & purificación , Ratones , Proteínas de Unión a Poli-ADP-Ribosa , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/enzimología , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA