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

Banco de datos
Tipo del documento
Intervalo de año de publicación
2.
J Cell Biol ; 127(6 Pt 1): 1527-36, 1994 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-7798309

RESUMEN

Calmodulin (CaM) potentiates Ca(2+)-dependent signaling pathways in both the cytoplasm and nucleus. We have investigated the mechanism of CaM nuclear transport using tissue culture cell microinjection and a permeabilized cell import assay. The inhibition of CaM import by the translocation inhibitor wheat germ agglutinin (WGA) and by chilling, indicates that CaM import is facilitated, but because ATP depletion does not affect CaM import, the mechanism does not appear to be active. Chilling and WGA arrest persist in ATP-depleted cells, indicating that CaM is not retained in the cytoplasm by an ATP-dependent mechanism. In permeabilized cells, both Ca(2+)-CaM and Ca(2+)-free CaM are sensitive to extract-dependent WGA and chilling import inhibition. Titration experiments in microinjected and permeabilized cells indicate that a saturable cytosolic factor(s) mediates chilling and WGA arrest.


Asunto(s)
Calmodulina/metabolismo , Compartimento Celular , Núcleo Celular/metabolismo , Adenosina Trifosfato/metabolismo , Transporte Biológico/efectos de los fármacos , Calbindinas , Permeabilidad de la Membrana Celular , Células Cultivadas , Frío , Histocitoquímica , Microinyecciones , Microscopía Fluorescente , Fragmentos de Péptidos/metabolismo , Proteína G de Unión al Calcio S100/metabolismo , Troponina/metabolismo , Troponina C , Aglutininas del Germen de Trigo/farmacología
4.
Curr Biol ; 7(5): 308-15, 1997 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-9133345

RESUMEN

BACKGROUND: Classical cadherin-based cellular adhesion is mediated by a multicomponent protein complex that links the adhesive binding activity of the cadherin ectodomain to the actin cytoskeleton. Despite the importance of cadherins in morphogenesis and development, we know very little about how cells determine and alter cadherin adhesive strength. In this study, we sought to identify specific cellular mechanisms that modulate cadherin function by studying adhesion between cells transfected with Xenopus C-cadherin mutant molecules and substrata coated with the purified ectodomain of C-cadherin. RESULTS: Using the FKBP-FK1012 protein oligomerization system, we found that forced clustering, in cells, of cadherin mutants lacking the cytoplasmic tail significantly increased cellular adhesive strength. Therefore, redistribution of the adhesive binding sites of cells into clusters can influence adhesion independently of other protein interactions mediated by the cadherin cytoplasmic tail. Furthermore, cells transfected with full-length C-cadherin demonstrated dynamic changes in adhesion over time that correlated with clustering but not with changes in the surface expression of C-cadherin or in the composition of the cadherin-catenin complex. The cytoplasmic tail was, however, necessary for clustering of wild-type cadherin. CONCLUSIONS: These studies directly demonstrate a fundamental role for lateral clustering in cadherin function. The distribution of cadherin binding sites presented at the cell surface, a cellular property which is regulated by the cadherin cytoplasmic tail, is an important mechanism which modulates cellular adhesion independently of cytoskeletal activity or signalling.


Asunto(s)
Cadherinas/fisiología , Adhesión Celular , Animales , Células CHO , Cadherinas/química , Cadherinas/aislamiento & purificación , Proteínas Portadoras/metabolismo , Membrana Celular/fisiología , Cricetinae , Cartilla de ADN , Proteínas de Unión al ADN/metabolismo , Proteínas de Choque Térmico/metabolismo , Cinética , Reacción en Cadena de la Polimerasa , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/metabolismo , Tacrolimus , Proteínas de Unión a Tacrolimus , Transfección , Xenopus
5.
Eur J Cancer ; 43(12): 1833-41, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17627812

RESUMEN

Loss of caspase-8 expression - which has been demonstrated in a subset of Medulloblastoma (MB) - might block important apoptotic signalling pathways and therefore contribute to treatment resistance. In this study, IFN-gamma mediated up-regulation of caspase-8 in human MB cells was found to result in chemosensitization to cisplatin, doxorubicin and etoposide, and sensitisation to radiation. These effects were more prominent in D425 and D341 MB cells (low basal caspase-8 expression) when compared to DAOY MB cells (high basal caspase-8 expression). IFN-gamma mediated chemosensitization and radiosensitization effects were reduced by treatment with the caspase-8 specific inhibitor z-IETD-fmk. Treatment of IFN-gamma resulted in activation of STAT1 in DAOY MB cells and to a lesser extent in D425, but not in D341, indicating that IFN-gamma acts in MB cells through STAT1-dependent and -independent signalling pathways. Taken together, our results demonstrate that IFN-gamma mediated restoration of caspase-8 in MB cells might enhance apoptotic pathways relevant to the response to chemo- and radiotherapy.


Asunto(s)
Antineoplásicos/farmacología , Caspasa 8/metabolismo , Neoplasias Cerebelosas/metabolismo , Interferón gamma/farmacología , Meduloblastoma/metabolismo , Fármacos Sensibilizantes a Radiaciones/farmacología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Línea Celular Tumoral , Neoplasias Cerebelosas/tratamiento farmacológico , Neoplasias Cerebelosas/radioterapia , Niño , Femenino , Humanos , Masculino , Meduloblastoma/tratamiento farmacológico , Meduloblastoma/radioterapia , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Regulación hacia Arriba
6.
Mol Cell Biol ; 15(2): 661-70, 1995 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-7823935

RESUMEN

It was previously reported that the phosphorylation of three proteins of 36, 40 to 42, and 50 kDa by casein kinase 2 is inhibited by calmodulin in nuclear extracts from rat liver cells (R. Bosser, R. Aligué, D. Guerini, N. Agell, E. Carafoli, and O. Bachs, J. Biol. Chem. 268:15477-15483, 1993). By immunoblotting, peptide mapping, and endogenous phosphorylation experiments, the 36- and 40- to 42-kDa proteins have been identified as the A2 and C proteins, respectively, of the heterogeneous nuclear ribonucleoprotein particles. To better understand the mechanism by which calmodulin inhibits the phosphorylation of these proteins, they were purified by using single-stranded DNA chromatography, and the effect of calmodulin on their phosphorylation by casein kinase 2 was analyzed. Results revealed that whereas calmodulin inhibited the phosphorylation of purified A2 and C proteins in a Ca(2+)-dependent manner, it did not affect the casein kinase 2 phosphorylation of a different protein substrate, i.e., beta-casein. These results indicate that the effect of calmodulin was not on casein kinase 2 activity but on specific protein substrates. The finding that the A2 and C proteins can bind to a calmodulin-Sepharose column in a Ca(2+)-dependent manner suggests that this association could prevent the phosphorylation of the proteins by casein kinase 2. Immunoelectron microscopy studies have revealed that such interactions could also occur in vivo, since calmodulin and A2 and C proteins colocalize on the ribonucleoprotein particles in rat liver cell nuclei.


Asunto(s)
Calmodulina/farmacología , Núcleo Celular/metabolismo , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B , Hígado/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Ribonucleoproteínas/metabolismo , Adenosina Trifosfato/metabolismo , Secuencia de Aminoácidos , Animales , Quinasa de la Caseína II , Cromatografía de Afinidad , Guanosina Trifosfato/metabolismo , Células HeLa , Ribonucleoproteínas Nucleares Heterogéneas , Humanos , Masculino , Microscopía Inmunoelectrónica , Datos de Secuencia Molecular , Fragmentos de Péptidos/química , Fragmentos de Péptidos/aislamiento & purificación , Mapeo Peptídico , Fosforilación , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Ratas , Ratas Sprague-Dawley , Ribonucleoproteínas/análisis , Ribonucleoproteínas/aislamiento & purificación , Homología de Secuencia de Aminoácido , Serina Endopeptidasas , Especificidad por Sustrato
7.
Cancer Res ; 61(22): 8203-10, 2001 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-11719451

RESUMEN

Activation of the phosphatidylinositol 3'-kinase (PI3K)/Akt survival pathway protects against apoptotic stress stimuli. Therefore, compounds that down-regulate this pathway are of clinical interest for single and combined anticancer treatment modalities. Here we demonstrate that the cytotoxic effect of the protein kinase C (PKC)-inhibitor N-benzoylated staurosporine (PKC412) is mediated via the PI3K/Akt pathway. Dose-dependent down-regulation of the proliferative activity, activation of the apoptotic machinery, and cell killing by PKC412 (0-1 microM) in Rat1a-fibroblasts and H-ras-oncogene-transformed fibroblasts correlated with a decrease of Akt phosphorylation and a reduced phosphorylation of the endogenous Akt-substrate GSK3-alpha. Expression of the dominant-active myristoylated form of Akt abrogated this cytotoxic effect of PKC412. Experiments with Apaf-1-deficient cells revealed that PKC412-induced cytotoxicity depends on an intact apoptosome but that the decrease of Akt phosphorylation is not attributable to apoptosis execution. Comparative experiments indicate that PKC412 and the parent-compound staurosporine down-regulate this survival pathway upstream or at the level of Akt but by a different mechanism than the PI3K-inhibitor LY294002. Furthermore, inhibition of this pathway by PKC412 is relevant for sensitization to ionizing radiation. These results demonstrate the specific role of this signaling pathway for the PKC412-mediated down-regulation of an apoptotic threshold and its cytotoxicity.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Fosfatidilinositol 3-Quinasas/fisiología , Proteína Quinasa C/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas/fisiología , Transducción de Señal/efectos de los fármacos , Estaurosporina/análogos & derivados , Estaurosporina/farmacología , Animales , Línea Celular Transformada , Transformación Celular Neoplásica/genética , Cromonas/farmacología , Regulación hacia Abajo/efectos de los fármacos , Genes ras/fisiología , Humanos , Morfolinas/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Ratas , Transducción de Señal/fisiología , Células Tumorales Cultivadas
8.
Cancer Res ; 61(2): 732-8, 2001 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-11212276

RESUMEN

The cellular response to ionizing radiation is governed by the DNA-damage recognition process but is also modulated by cytoplasmic signal transduction cascades that are part of the cellular stress response. Growth-promoting protein kinase C activity antagonizes irradiation-induced cell death, and, therefore, protein kinase C inhibitors might be potent radiosensitizers. The antiproliferative and radiosensitizing effect of the novel N-benzoylated staurosporine analogue PKC412 was tested in vitro against genetically defined p53-wild type (+/+) and p53-deficient (-/-) murine fibrosarcoma cells and in vivo against radioresistant p53-/- murine fibrosarcoma and human colon adenocarcinoma tumor xenograft (SW480, p53-mutated). PKC412 sensitized both p53+/+ and p53-/- tumor cells in vitro and in vivo for treatment with ionizing radiation but with a different mechanism of radiosensitization depending on the p53 status. In p53+/+, cells combined treatment with PKC412 and ionizing radiation drastically induced apoptotic cell death, whereas no apoptosis induction could be observed in p53-deficient cells in vitro and in histological tumor sections. Combined treatment resulted in an increased G2 cell cycle distribution in p53-/- cells at PKC412 concentrations that did not alter cell cycle distribution when applied alone. In vivo, a minimal treatment regimen during 4 consecutive days of PKC412 (4 x 100 mg/kg) in combination with ionizing radiation (4 x 3 Gy) exerted a substantial tumor growth delay for both p53-disfunctional tumor xenografts and showed that the clinically relevant protein kinase C inhibitor PKC412 is a promising new radiosensitizer with a potentially broad therapeutic window.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Proteína Quinasa C/antagonistas & inhibidores , Estaurosporina/análogos & derivados , Estaurosporina/farmacología , Proteína p53 Supresora de Tumor/fisiología , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Células Cultivadas , Terapia Combinada , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Genotipo , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/patología , Neoplasias Experimentales/radioterapia , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/efectos de la radiación , Proteína p53 Supresora de Tumor/genética , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Phys Med Biol ; 61(24): 8736-8749, 2016 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-27893446

RESUMEN

This study aimed to identify a set of stable radiomic parameters in CT perfusion (CTP) maps with respect to CTP calculation factors and image discretization, as an input for future prognostic models for local tumor response to chemo-radiotherapy. Pre-treatment CTP images of eleven patients with oropharyngeal carcinoma and eleven patients with non-small cell lung cancer (NSCLC) were analyzed. 315 radiomic parameters were studied per perfusion map (blood volume, blood flow and mean transit time). Radiomics robustness was investigated regarding the potentially standardizable (image discretization method, Hounsfield unit (HU) threshold, voxel size and temporal resolution) and non-standardizable (artery contouring and noise threshold) perfusion calculation factors using the intraclass correlation (ICC). To gain added value for our model radiomic parameters correlated with tumor volume, a well-known predictive factor for local tumor response to chemo-radiotherapy, were excluded from the analysis. The remaining stable radiomic parameters were grouped according to inter-parameter Spearman correlations and for each group the parameter with the highest ICC was included in the final set. The acceptance level was 0.9 and 0.7 for the ICC and correlation, respectively. The image discretization method using fixed number of bins or fixed intervals gave a similar number of stable radiomic parameters (around 40%). The potentially standardizable factors introduced more variability into radiomic parameters than the non-standardizable ones with 56-98% and 43-58% instability rates, respectively. The highest variability was observed for voxel size (instability rate >97% for both patient cohorts). Without standardization of CTP calculation factors none of the studied radiomic parameters were stable. After standardization with respect to non-standardizable factors ten radiomic parameters were stable for both patient cohorts after correction for inter-parameter correlations. Voxel size, image discretization, HU threshold and temporal resolution have to be standardized to build a reliable predictive model based on CTP radiomics analysis.


Asunto(s)
Imagen de Perfusión/métodos , Tomografía Computarizada por Rayos X/métodos , Volumen Sanguíneo , Carcinoma de Pulmón de Células no Pequeñas/diagnóstico por imagen , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/fisiopatología , Humanos , Neoplasias Pulmonares/diagnóstico por imagen , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/fisiopatología , Neoplasias Orofaríngeas/diagnóstico por imagen , Neoplasias Orofaríngeas/patología , Neoplasias Orofaríngeas/fisiopatología , Pronóstico , Carga Tumoral
10.
Diabetes ; 50(9): 2105-13, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11522678

RESUMEN

Several studies support the concept of a diabetic cardiomyopathy in the absence of discernible coronary artery disease, although its mechanism remains poorly understood. We investigated the role of glucose and palmitic acid on cardiomyocyte apoptosis and on the organization of the contractile apparatus. Exposure of adult rat cardiomyocytes for 18 h to palmitic acid (0.25 and 0.5 mmol/l) resulted in a significant increase of apoptotic cells, whereas increasing glucose concentration to 33.3 mmol/l for up to 8 days had no influence on the apoptosis rate. However, both palmitic acid and elevated glucose concentration alone or in combination had a dramatic destructive effect on the myofibrillar apparatus. The membrane-permeable C2-ceramide but not the metabolically inactive C2-dihydroceramide enhanced apoptosis of cardiomyocytes by 50%, accompanied by detrimental effects on the myofibrils. The palmitic acid-induced effects were impaired by fumonisin B1, an inhibitor of ceramide synthase. Sphingomyelinase, which activates the catabolic pathway of ceramide by metabolizing sphingomyeline to ceramide, did not adversely affect cardiomyocytes. Palmitic acid-induced apoptosis was accompanied by release of cytochrome c from the mitochondria. Aminoguanidine did not prevent glucose-induced myofibrillar degeneration, suggesting that formation of nitric oxide and/or advanced glycation end products play no major role. Taken together, these results suggest that in adult rat cardiac cells, palmitic acid induces apoptosis via de novo ceramide formation and activation of the apoptotic mitochondrial pathway. Conversely, glucose has no influence on adult cardiomyocyte apoptosis. However, both cell nutrients promote degeneration of myofibrils. Thus, gluco- and lipotoxicity may play a central role in the development of diabetic cardiomyopathy.


Asunto(s)
Apoptosis/efectos de los fármacos , Glucosa/farmacología , Corazón/fisiología , Miofibrillas/efectos de los fármacos , Ácido Palmítico/farmacología , Animales , Células Cultivadas , Ceramidas/fisiología , Grupo Citocromo c/metabolismo , Femenino , Corazón/efectos de los fármacos , Miocardio/citología , Miocardio/metabolismo , Miofibrillas/patología , Miofibrillas/fisiología , Ácidos Oléicos/farmacología , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología
11.
Chem Biol ; 1(3): 163-72, 1994 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-9383386

RESUMEN

BACKGROUND: The T-cell receptor (TCR) signaling pathway is initiated by regulated association of TCR chains, including the zeta chain. A recently reported method for inducing the dimerization or oligomerization of targeted proteins in cells used the TCR pathway as a test system. In cells transfected with cDNA encoding MZF3E, a chimeric receptor comprising the intracellular domain of the zeta chain and three copies of FK506-binding protein (FKBP), low concentrations of a synthetic dimer of the natural product FK506 (FK1012) activated the expression of reporter genes. We set out to examine the signaling pathway initiated by FK1012. RESULTS: We characterized the effect of FK1012 on MZF3E and a second chimeric receptor, MZF1E, which contains the zeta chain and one copy of FKBP. Only MZF3E gave FK1012-activated signaling, as shown by an increase in the kinase activity associating with MZF3E, and the appearance of specific phosphotyrosine-containing proteins. Signaling required localization of MZF3E to the inner plasma membrane, and activation of gene transcription in response to FK1012 was dependent on the protein phosphatase calcineurin and the transcriptional activator NF-AT. Some signaling events in the pathway had different kinetics when activated by MZF3E instead of the TCR, however. An unexpected requirement for the prolonged activation of calcineurin was observed. CONCLUSIONS: Synthetic dimerizers can be used to gain control over cellular processes that require the association of specific intracellular proteins. The TCR signaling pathway was selected as an initial test system; we show here that one can indeed activate this signaling pathway by inducing the oligomerization of the cytoplasmic tail of the zeta chain with the cell-permeable reagent FK1012.


Asunto(s)
Receptores de Antígenos de Linfocitos T/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Tacrolimus/análogos & derivados , Animales , Células Cultivadas , Humanos , Receptores de Antígenos de Linfocitos T/genética , Transducción de Señal/genética , Tacrolimus/síntesis química , Tacrolimus/farmacología , Transfección
12.
Curr Med Chem Anticancer Agents ; 3(1): 35-46, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12678913

RESUMEN

Cell cycle checkpoints are activated in response to DNA-damage to ensure that accurate copies of the cellular genome are passed on to the next generation and to avoid replication and segregation of damaged DNA. These cellular control systems can be overcome by combining conventional DNA-damaging agents with compounds that target the cell cycle regulatory pathways, to enhance cytotoxicity. Tumor cells often comprise a corrupted G(1) cell cycle checkpoint while the G(2) cell cycle checkpoint is still intact. This review describes the concept of G(2) checkpoint abrogation with recognized (methylxanthines, UCN-01) and novel G(2) checkpoint abrogators to potentiate the cytotoxicity of DNA-damaging drugs and ionizing radiation. It illustrates the potential of G(2) checkpoint abrogators to preferentially sensitize p53-mutated, treatment resistant tumor cells for genotoxic treatment. Identification of the targets of caffeine and UCN-01 to be key-players of the G(2) checkpoint (ATM/ATR and Chk1, respectively) promoted the search for novel inhibitors of this checkpoint. Even though a direct causal link between G(2) checkpoint abrogation and chemo-/radiosensitization is difficult to prove the multitude of these novel compounds validate that inhibition of critical elements of the G(2) checkpoint (ATM/ATR-Chk1/Chk2-CDC25C-cascade) potentiates the cytotoxicity of DNA-damaging agents.


Asunto(s)
Antineoplásicos/farmacología , Proteínas de Ciclo Celular/antagonistas & inhibidores , Daño del ADN/efectos de los fármacos , Fase G2/efectos de los fármacos , Alcaloides , Animales , Antineoplásicos/química , Cafeína/química , Cafeína/farmacología , Supervivencia Celular/efectos de los fármacos , Terapia Combinada , Daño del ADN/efectos de la radiación , Sinergismo Farmacológico , Humanos , Estaurosporina/análogos & derivados , Xantinas/química , Xantinas/farmacología
13.
Curr Med Chem Anticancer Agents ; 2(6): 727-42, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12678723

RESUMEN

DNA double strand breaks are the pivotal cellular damage induced by ionizing radiation. A plethora of molecular and cellular processes are activated as part of the cellular stress response that result in cell cycle arrest and induction of the DNA-repair machinery to restore the damage of DNA or to activate a cell death program. However ionizing radiation also initiates signal transduction cascades that are generated at cellular sites distant from and independent of DNA-damage. These signaling processes are similar to hormone activated growth factor receptor controlled signal transduction cascades and represent interesting targets for anticancer treatment modalities combining ionizing radiation with molecular defined pharmacological compounds. Activation of these signal transduction cascades upon irradiation or upregulation of growth factor mediated pathways due to oncogene-transformation often contribute to an acquired or inherent treatment resistance in malignant cells. Therefore pharmacological compounds inhibiting specific key-entities of these signal transduction cascades potentially sensitize for radiation induced cell death. Here we describe current preclinical concepts of combined treatment strategies with locoregional-applied ionizing radiation and molecular defined signal transduction inhibitors to overcome a high treatment threshold in tumor cells.


Asunto(s)
Neoplasias/radioterapia , Fármacos Sensibilizantes a Radiaciones/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Humanos , Ligandos , Neoplasias/tratamiento farmacológico , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Radioterapia/efectos adversos , Radioterapia/métodos
14.
Int J Radiat Oncol Biol Phys ; 49(2): 561-7, 2001 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-11173155

RESUMEN

In many human hematologic and solid malignancies, intrinsic or acquired treatment resistance remains a major obstacle for successful cancer therapy. The molecular understanding of how tumor cells respond to chemotherapy and ionizing radiation is rapidly evolving. Induction of programmed cell death, apoptosis, is one important strategy for successful cancer therapy. This has been shown convincingly for oncogene-transformed normal cells as well as tumor cells of lymphoid origin. However, the relevance of apoptosis in solid human malignancies is less clear. Loss of apoptosis might be linked to specific mutations in the often tissue-specific apoptotic pathways due to aberrations in the stress-related signal transduction cascades. Restoration of a dysfunctional apoptotic program in cancer tissue where apoptosis has been identified as an important mechanism for tissue homeostasis is one rational approach for innovative cancer therapy. In this review, we focus on the relevance of the tumor suppressor p53 for apoptosis-induction and successful cancer therapy outlining the importance of an intact caspase machinery for apoptosis execution. Strategies are discussed to overcome treatment resistance and a high apoptotic threshold in human malignancies where apoptosis is the dominant mode of cell death and the status of p53 is an important determinant for apoptosis induction.


Asunto(s)
Apoptosis/genética , Caspasas/fisiología , Genes p53/fisiología , Terapia Genética/métodos , Vectores Genéticos/uso terapéutico , Proteínas de Neoplasias/fisiología , Neoplasias/terapia , Adenoviridae/genética , Animales , Apoptosis/fisiología , Ciclo Celular/fisiología , Reparación del ADN , Activación Enzimática , Inhibidores Enzimáticos/uso terapéutico , Genes p53/genética , Vectores Genéticos/genética , Humanos , Ratones , Ratones Noqueados , Neoplasias/genética , Neoplasias/radioterapia , Estaurosporina/uso terapéutico
15.
Oncogene ; 30(25): 2823-35, 2011 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-21317922

RESUMEN

Medulloblastoma (MB) is the most common malignant brain tumor in children. It is known that overexpression and/or amplification of the MYC oncogene is associated with poor clinical outcome, but the molecular mechanisms and the MYC downstream effectors in MB remain still elusive. Besides contributing to elucidate how progression of MB takes place, most importantly, the identification of novel MYC-target genes will suggest novel candidates for targeted therapy in MB. A group of 209 MYC-responsive genes was obtained from a complementary DNA microarray analysis of a MB-derived cell line, following MYC overexpression and silencing. Among the MYC-responsive genes, we identified the members of the bone morphogenetic protein (BMP) signaling pathway, which have a crucial role during the development of the cerebellum. In particular, the gene BMP7 was identified as a direct target of MYC. A positive correlation between MYC and BMP7 expression was documented by analyzing two distinct sets of primary MB samples. Functional studies in vitro using a small-molecule inhibitor of the BMP/SMAD signaling pathway reproduced the effect of the small interfering RNA-mediated silencing of BMP7. Both approaches led to a block of proliferation in a panel of MB cells and to inhibition of SMAD phosphorylation. Altogether, our findings indicate that high MYC levels drive BMP7 overexpression, promoting cell survival in MB cells. This observation suggests the potential relevance of targeting the BMP/SMAD pathway as a novel therapeutic approach for the treatment of childhood MB.


Asunto(s)
Proteína Morfogenética Ósea 7/genética , Supervivencia Celular/genética , Neoplasias Cerebelosas/genética , Genes myc , Meduloblastoma/genética , Western Blotting , Neoplasias Cerebelosas/patología , Niño , Ensayo de Inmunoadsorción Enzimática , Silenciador del Gen , Humanos , Meduloblastoma/patología , Fosforilación , Proteínas Smad/metabolismo
17.
Gene Ther ; 14(15): 1143-51, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17495946

RESUMEN

Standard treatment for glioblastoma multiforme and other brain tumors consists of surgical resection followed by combined radio-/chemotherapy. However, radiation resistance of tumor cells limits the success of this treatment, and the tumors invariably recur. Therefore, the selective inhibition of molecular mediators of radiation resistance may provide therapeutic benefit to the patient. One of these targets is the Rad51 protein, which is a key component of the homologous recombinational repair of DNA double-strand breaks. Here, we investigated whether post-transcriptional silencing of Rad51 by herpes simplex virus-type 1 (HSV-1) amplicon vector-mediated short interfering RNA expression can enhance the antitumor effect of radiation therapy. We demonstrate that these vectors specifically and efficiently inhibited the radiation-induced recruitment of Rad51 into nuclear foci in human glioma cells. The combination of vector-mediated silencing of Rad51 expression and treatment with ionizing radiation resulted in a pronounced reduction of the survival of human glioma cells in culture. In athymyc mice, a single intratumoral injection of Rad51-specific HSV-1 amplicon vector followed by a single radiation treatment resulted in a significant decrease in tumor size. In control animals, including mice that received an intratumoral injection of Rad51-specific amplicon vector but no radiation treatment, the tumor sizes increased.


Asunto(s)
Neoplasias Encefálicas/terapia , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Glioma/terapia , Herpesvirus Humano 1/genética , Recombinasa Rad51/antagonistas & inhibidores , Animales , Apoptosis , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/radioterapia , Línea Celular Tumoral , Terapia Combinada , Femenino , Expresión Génica , Silenciador del Gen , Vectores Genéticos/genética , Glioblastoma/terapia , Glioma/patología , Glioma/radioterapia , Humanos , Inyecciones Intralesiones , Ratones , Ratones Desnudos , Interferencia de ARN , ARN Interferente Pequeño/genética , Recombinasa Rad51/genética , Tolerancia a Radiación/fisiología , Resultado del Tratamiento
18.
Cell Growth Differ ; 10(3): 141-6, 1999 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10099827

RESUMEN

Negative regulation of E2F-1 DNA binding function by cyclin A kinase represents part of an S-phase checkpoint control system that, when activated, leads to apoptosis. In this study, we examined the cellular sensitivity and resistance of isogenic mouse fibrosarcoma cell lines, differing primarily in their p53 status, to ectopic expression of wild-type (wt) E2F-1 and cyclin A kinase binding-defective mutants of it. We found that E2F-1 (wt) potently affected the survival of p53+/+ tumor cells but not that of p53-/- cells. In contrast, expression of cyclin A kinase binding-defective E2F-1 species interfered with cell survival of fibrosarcoma cells irrespective of their p53 status. Finally, expression of E2F-1 (wt) in p53-/- fibrosarcoma cells enhanced the cytotoxic effect of ionizing radiation in vitro and in vivo in a mouse tumor model. These results suggest that E2F-1-dependent activation of an S-phase checkpoint is p53 independent and that E2F-1 possesses radiosensitizing properties in the absence of p53.


Asunto(s)
Proteínas Portadoras , Proteínas de Ciclo Celular , Proteínas de Unión al ADN , Fibrosarcoma/metabolismo , Genes p53 , Proteínas Quinasas/metabolismo , Factores de Transcripción/metabolismo , Animales , Línea Celular Transformada , Supervivencia Celular , Factores de Transcripción E2F , Factor de Transcripción E2F1 , Fibrosarcoma/genética , Expresión Génica , Ratones , Ratones Desnudos , Inhibidores de la Síntesis de la Proteína/metabolismo , Puromicina/metabolismo , Tolerancia a Radiación/fisiología , Proteína 1 de Unión a Retinoblastoma , Factores de Tiempo , Factor de Transcripción DP1 , Factores de Transcripción/farmacología , Proteína p53 Supresora de Tumor/deficiencia
19.
Br J Cancer ; 80(5-6): 693-8, 1999 May.
Artículo en Inglés | MEDLINE | ID: mdl-10360645

RESUMEN

p53 mutations are among the most common genetic alterations in human cancer and are frequently described in intrinsic or acquired radio- and chemotherapy resistance. Radiation-induced cell kill is not only mediated by DNA damage but also by the activation of signal transduction cascades generated at the plasma membrane like the sphingomyelin pathway. We used genetically defined wild-type p53 or p53-deficient mouse fibrosarcoma cells to investigate the p53-dependence of tumour response upon activation of the sphingomyelin pathway. Treatment of the tumour cells with neutral sphingomyelinase drastically reduced the amount of wild-type p53 fibrosarcoma cell proliferation over 72 h in a clear dose-response (0.2-1.0 U ml(-1) nSMase). Sphingomyelinase had no effect on cell proliferation in tumour cells lacking p53. Similarly, cell proliferation was abolished by C2-ceramide (5-20 microM) only in wild-type p53 cells. FACS-analysis revealed that C2-ceramide induced massive p53-dependent apoptosis (40-50% after 12-24 h) and cell cycle analysis showed a transient G1 arrest in p53-deficient tumour cells 12-24 h after C2-ceramide exposure. These results suggest that ceramide-induced apoptosis in tumour cells can be dependent on the status of p53 and imply that p53 is also important for stress-induced apoptotic signal transduction cascades generated at the plasma membrane.


Asunto(s)
Apoptosis/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Fibrosarcoma/genética , Fibrosarcoma/patología , Esfingosina/análogos & derivados , Proteína p53 Supresora de Tumor/fisiología , Animales , Apoptosis/genética , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Fibrosarcoma/tratamiento farmacológico , Inhibidores de Crecimiento/farmacología , Ratones , Ratones Noqueados , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Esfingomielina Fosfodiesterasa/farmacología , Esfingomielinas/fisiología , Esfingosina/farmacología , Células Tumorales Cultivadas
20.
Biochemistry ; 28(10): 4253-8, 1989 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-2548572

RESUMEN

The primary structure of a region of the erythrocyte plasma membrane calcium pump which is phosphorylated by the cAMP-dependent protein kinase has been determined. The sequence is A-P-T-K-R-N-S-S(P)-P-P-P-S-P-D. The site is located between the calmodulin binding domain and the C-terminus of the ATPase. The ATPase is phosphorylated only at this site by the cAMP-dependent protein kinase, and the phosphorylation is inhibited by calmodulin. The effect of the phosphorylation is to decrease the Km for Ca2+ of the purified ATPase from about 10 microM to about 1.4 microM and to increase the Vmax of ATP hydrolysis about 2-fold.


Asunto(s)
Canales de Calcio/metabolismo , Membrana Celular/metabolismo , AMP Cíclico/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Membrana Eritrocítica/metabolismo , Humanos , Técnicas In Vitro , Cinética , Datos de Secuencia Molecular , Estructura Molecular , Fosforilación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA