Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
Breast Cancer Res Treat ; 149(2): 385-94, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25528024

RESUMEN

Recently, we observed that telomeres of BRCA1/2 mutation carriers were shorter than those of controls or sporadic breast cancer patients, suggesting that mutations in these genes might be responsible for this event. Given the contradictory results reported in the literature, we tested whether other parameters, such as chemotherapy, could be modifying telomere length (TL). We performed a cross-sectional study measuring leukocyte TL of 266 sporadic breasts cancer patients treated with first-line chemotherapy, with a median follow-up of 240 days. Additionally, we performed both cross-sectional and longitudinal studies in a series of 236 familial breast cancer patients that included affected and non-affected BRCA1/2 mutation carriers. We have measured in leukocytes from peripheral blood: the TL, percentage of short telomeres (<3 kb), telomerase activity levels and the annual telomere shortening speed. In sporadic cases we found that chemotherapy exerts a transient telomere shortening effect (around 2 years) that varies depending on the drug combination. In familial cases, only patients receiving treatment were associated with telomere shortening but they recovered normal TL after a period of 2 years. Chemotherapy affects TL and should be considered in the studies that correlate TL with disease susceptibility.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Mama/genética , Telómero/genética , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias de la Mama/tratamiento farmacológico , Estudios de Casos y Controles , Estudios Transversales , Femenino , Genes BRCA1 , Genes BRCA2 , Humanos , Estudios Longitudinales , Persona de Mediana Edad , Mutación , Factores de Riesgo , Telómero/metabolismo , Acortamiento del Telómero , Adulto Joven
2.
Apoptosis ; 18(3): 347-60, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23271172

RESUMEN

Our results demonstrate that the addition of cisplatin after paclitaxel-induced mitotic arrest was more effective than individual treatment on gastric adenocarcinoma cells (MKN45). However, the treatment did not induce benefits in cells derived from lymph node metastasis (ST2957). Time-lapse microscopy revealed that cell death was caused by mitotic catastrophe and apoptosis induction, as the use of the caspase inhibitor z-VAD-fmk decreased cell death. We propose that the molecular mechanism mediating this cell fate is a slippage suffered by these cells, given that our Western blot (WB) analysis revealed premature cyclin B degradation. This resulted in the cell exiting from mitosis without undergoing DNA damage repair, as demonstrated by the strong phosphorylation of H2AX. A comet assay indicated that DNA repair was impaired, and Western blotting showed that the Chk2 protein was degraded after sequential treatment (paclitaxel-cisplatin). Based on these results, the modulation of cell death during mitosis may be an effective strategy for gastric cancer therapy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Apoptosis/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , Mitosis/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/efectos de los fármacos , Neoplasias Gástricas/tratamiento farmacológico , Clorometilcetonas de Aminoácidos/farmacología , Línea Celular Tumoral , Quinasa de Punto de Control 2 , Cisplatino/uso terapéutico , Ciclina B/metabolismo , Humanos , Metástasis Linfática/patología , Paclitaxel , Proteínas Serina-Treonina Quinasas/metabolismo , Taxoides/uso terapéutico
3.
Blood Cells Mol Dis ; 49(3-4): 140-6, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22664374

RESUMEN

Dyskeratosis congenita (DC) is a rare inherited bone-marrow failure syndrome with high clinical heterogeneity. Cells derived from DC patients present short telomeres at early ages, as a result of mutations in genes encoding components of the telomerase complex (DKC1, TERC, TERT, NHP2 and NOP10), or the shelterin complex (TINF2). However, mutations have been identified only in around 50% of the cases, indicating that other genes could be involved in the development of this disease. Indeed, mutations in TCBA1 or chromosome segment C16orf57 have been described recently. We have used HRM technology to perform genetic analysis in the above mentioned genes, in Spanish patients showing both, some clinical features of DC and short telomeres. The mutations have been identified by PCR amplification of DC genes followed by high resolution melting (HRM) and direct DNA sequencing analysis. We have identified seven new families with DC, three with X-linked DC and four with autosomal dominant DC, in which we have found two novel mutations in DKC1 (p.His68Arg and p.Lys390del) and four novel mutations in TERT gene (p.Pro530Leu, p.Arg698Trp, p.Arg971His and p.Arg698Gln). The results show that the use of HRM analysis enables a rapid and inexpensive identification of mutations in dyskeratosis congenita associated genes.


Asunto(s)
Proteínas de Ciclo Celular/genética , Disqueratosis Congénita/genética , Proteínas Nucleares/genética , Análisis de Secuencia de ADN/métodos , Telomerasa/genética , Adolescente , Adulto , Médula Ósea/metabolismo , Médula Ósea/patología , Niño , Preescolar , Disqueratosis Congénita/diagnóstico , Disqueratosis Congénita/patología , Femenino , Humanos , Masculino , Datos de Secuencia Molecular , Mutación , Desnaturalización de Ácido Nucleico , Reacción en Cadena de la Polimerasa , Telómero/patología , Población Blanca
6.
Oncogene ; 25(23): 3335-45, 2006 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-16462770

RESUMEN

Non-small-cell lung cancer (NSCLC) represents the most frequent and therapy-refractive sub-class of lung cancer. Improving apoptosis induction in NSCLC represents a logical way forward in treating this tumor. Cisplatin, a commonly used therapeutic agent in NSCLC, induces activation of N-terminal-c-Jun kinase (JNK) that, in turn, mediates induction of apoptosis. In analysing surgical tissue samples of NSCLC, we found that expression of MKP1/CL100, a negative regulator of JNK, showed a strong nuclear staining for tumor cells, whereas, in normal bronchial epithelia, MKP1 was localized in the cytoplasm as well as in nuclei. In the NSCLC-derived cell lines H-460 and H-23, we found that MKP1 was constitutively expressed. Expressing a small-interfering RNA (siRNA) vector for MKP1 in H-460 cells resulted in a more efficient activation by cisplatin of JNK and p38 than in the parental cells, and this correlated with a 10-fold increase in sensitivity to cisplatin. A similar response was also observed in H-460 and H-23 cells when treated with the MKP1 expression inhibitor RO-31-8220. Moreover, expression of a siRNA-MKP2, an MKP1-related phosphatase, had no effect on H-460 cell viability response to cisplatin. Tumors induced by H-460 cells expressing MKP1 siRNA grew slower in nu(-)/nu(-) mice and showed more susceptibility to cisplatin than parental cells, and resulted in an impaired growth of the tumor in mice. On the other hand, overexpression of MKP1 in the H-1299 NSCLC-derived cell line resulted in further resistance to cisplatin. Overall, the results showed that inhibition of MKP1 expression contributes to a slow down in cell growth in mice and an increase of cisplatin-induced cell death in NSCLC. As such, MKP1 can be an attractive target in sensitizing cells to cisplatin to increase the effectiveness of the drug in treating NSCLC.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/patología , Proteínas de Ciclo Celular/fisiología , Proliferación Celular , Cisplatino/farmacología , Resistencia a Antineoplásicos/fisiología , Inhibidores de Crecimiento/fisiología , Proteínas Inmediatas-Precoces/fisiología , Neoplasias Pulmonares/patología , Fosfoproteínas Fosfatasas/fisiología , Proteínas Tirosina Fosfatasas/fisiología , Adulto , Anciano , Animales , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/enzimología , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/biosíntesis , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Fosfatasa 1 de Especificidad Dual , Femenino , Inhibidores de Crecimiento/antagonistas & inhibidores , Inhibidores de Crecimiento/biosíntesis , Humanos , Proteínas Inmediatas-Precoces/antagonistas & inhibidores , Proteínas Inmediatas-Precoces/biosíntesis , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/enzimología , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Fosfoproteínas Fosfatasas/antagonistas & inhibidores , Fosfoproteínas Fosfatasas/biosíntesis , Proteína Fosfatasa 1 , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores , Proteínas Tirosina Fosfatasas/biosíntesis , ARN Interferente Pequeño/farmacología
7.
Mol Cell Biol ; 10(8): 4110-5, 1990 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-2142513

RESUMEN

Mouse fibroblasts expressing a yeast proton-pumping ATPase show tumorigenic transformation (R. Perona, and R. Serrano, Nature (London) 334:438-440, 1988). By expressing site-directed mutations of the yeast ATPase with different levels of activity, a close correlation has been found between enzyme activity, tumorigenic transformation, and intracellular pH measured by weak-acid distribution. Fibroblasts expressing the yeast proton-pumping ATPase showed increased capability to grow at acidic pH and to resist lethal acidification mediated by reversal of the Na(+)-H+ antiporter. Measurements with microelectrodes in individual cells demonstrated electrical hyperpolarization and confirmed the increased pH of cells expressing yeast ATPase. These results indicate that the yeast enzyme expressed in mouse fibroblasts has electrogenic proton-pumping activity and that this activity deregulates fibroblast growth. This suggests a connection between the biophysical phenomena of proton transport, intracellular pH, and membrane potential and the biochemical regulatory circuits based on protein kinases and transcription factors.


Asunto(s)
Transformación Celular Neoplásica , ATPasas de Translocación de Protón/genética , Saccharomyces cerevisiae/genética , Transfección , Animales , Secuencia de Bases , Células Cultivadas , Replicación del ADN , Fibroblastos/enzimología , Fibroblastos/metabolismo , Homeostasis , Concentración de Iones de Hidrógeno , Cinética , Potenciales de la Membrana , Ratones , Datos de Secuencia Molecular , Mutación , Sondas de Oligonucleótidos , ATPasas de Translocación de Protón/metabolismo , Saccharomyces cerevisiae/enzimología , Timidina/metabolismo
8.
Mol Biol Cell ; 12(10): 3282-94, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11598209

RESUMEN

Stats (signal transducers and activators of transcription) are latent cytoplasmic transcription factors that on a specific stimulus migrate to the nucleus and exert their transcriptional activity. Here we report a novel signaling pathway whereby RhoA can efficiently modulate Stat3 transcriptional activity by inducing its simultaneous tyrosine and serine phosphorylation. Tyrosine phosphorylation is exerted via a member of the Src family of kinases (SrcFK) and JAK2, whereas the JNK pathway mediates serine phosphorylation. Furthermore, cooperation of both tyrosine as well as serine phosphorylation is necessary for full activation of Stat3. Induction of Stat3 activity depends on the effector domain of RhoA and correlates with induction of both Src Kinase-related and JNK activities. Activation of Stat3 has biological implications. Coexpression of an oncogenic version of RhoA along with the wild-type, nontransforming Stat3 gene, significantly enhances its oncogenic activity on human HEK cells, suggesting that Stat3 is an essential component of RhoA-mediated transformation. In keeping with this, dominant negative Stat3 mutants or inhibition of its tyrosine or serine phosphorylation completely abrogate RhoA oncogenic potential. Taken together, these results indicate that Stat3 is an important player in RhoA-mediated oncogenic transformation, which requires simultaneous phosphorylation at both tyrosine and serine residues by specific signaling events triggered by RhoA effectors.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Proteínas de Unión al ADN/metabolismo , Quinasa 1 de Quinasa de Quinasa MAP , Proteínas Proto-Oncogénicas , Serina/metabolismo , Transactivadores/metabolismo , Tirosina/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Animales , Células CHO/metabolismo , Línea Celular/metabolismo , Cricetinae , Femenino , Fibroblastos/metabolismo , Humanos , Janus Quinasa 2 , Riñón/citología , Hígado/citología , Proteína Quinasa 8 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Ovario/citología , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Ratas , Ratas Endogámicas BUF , Factor de Transcripción STAT3 , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo , Familia-src Quinasas/metabolismo
9.
Clin Transl Oncol ; 9(10): 625-33, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17974523

RESUMEN

Chemotherapeutic agents and also radiotherapy trigger a series of signalling pathways in the cells that activate not only the apoptotic machinery but also cell survival pathways. Some of these pathways are also altered by genetic changes in specific type of tumours, and are different even between patients with the same tumour. Among these pathways, the majority of survival signals involve the ERK, AKT and nuclear factor-kappaB pathways and those related to cell death, which are driven mainly either by inhibition of such survival networks or by upregulation of the JNK/p38 MAP-kinases. Thus, the efficacy of a given chemotherapy appears as a result of the balance between cell death and survival pathways elicited in each individual tumour. Modulation of such survival pathways would help to increase the efficacy of chemotherapy. Different strategies based on conventional chemotherapy have been used in the past with modest success. The availability of new molecules such as inhibitors of survival pathways and the use of new technologies for the study of individual tumours would have a positive impact on patient survival.


Asunto(s)
Resistencia a Antineoplásicos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Transducción de Señal , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Humanos , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Factor de Transcripción ReIB/metabolismo
10.
Sci Rep ; 6: 21519, 2016 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-26867682

RESUMEN

Radiation has a limited but relevant role in the adjuvant therapy of gastric cancer (GC) patients. Since Chk1 plays a critical function in cellular response to genotoxic agents, we aimed to analyze the role of Chk1 in GC as a biomarker for radiotherapy resistance. We analyzed Chk1 expression in AGS and MKN45 human GC cell lines by RT-QPCR and WB and in a small cohort of human patient's samples. We demonstrated that Chk1 overexpression specifically increases resistance to radiation in GC cells. Accordingly, abrogation of Chk1 activity with UCN-01 and its expression with shChk1 increased sensitivity to bleomycin and radiation. Furthermore, when we assessed Chk1 expression in human samples, we found a correlation between nuclear Chk1 accumulation and a decrease in progression free survival. Moreover, using a luciferase assay we found that Chk1's expression is controlled by p53 and RB/E2F1 at the transcriptional level. Additionally, we present preliminary data suggesting a posttranscriptional regulation mechanism, involving miR-195 and miR-503, which are inversely correlated with expression of Chk1 in radioresistant cells. In conclusion, Chk1/microRNA axis is involved in resistance to radiation in GC, and suggests Chk1 as a potential tool for optimal stratification of patients susceptible to receive adjuvant radiotherapy after surgery.


Asunto(s)
Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/biosíntesis , Quimioradioterapia , Factor de Transcripción E2F1/metabolismo , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Proteínas de Unión a Retinoblastoma/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/terapia , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Bleomicina/farmacología , Línea Celular Tumoral , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/genética , Factor de Transcripción E2F1/genética , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de la radiación , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Humanos , Proteínas de Unión a Retinoblastoma/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Proteína p53 Supresora de Tumor/genética , Ubiquitina-Proteína Ligasas/genética
11.
Oncogene ; 16(4): 533-40, 1998 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-9484843

RESUMEN

Genotoxic stress triggers signalling pathways that either mediate cell killing or protection of affected cells. While induction of p53 is observed for most of the genotoxins, activation of MAPK/SAPK cascades is not a general response. The role of MAPK/SAPK activation on cell fate, seems to be dependent, in some systems, on the balanced response among both cascades. We have here examined the effect of cis and trans-DDP on the activation of ERK and JNK activities. While no significant induction of ERK was observed with the compounds, both of them are able to strongly activate JNK. Trans-DDP response is rapid and transient while the cis-DDP one is slow and persistent. In contrast with the observed nuclear translocation of JNK in response to U.V. light, none of the platinum compounds induces translocation, on the contrary, activation of JNK occurs in both the nuclear and cytoplasmic compartments. Inhibition of tyrosine phosphatases by orthovanadate pretreatment prolongs the time of JNK induction in response to both platinum compounds. The positive modulation of JNK activation correlates with an increase in toxicity that, for cis-DDP corresponds to a tenfold decrease in the IC50. A strong increase in MKP-1 levels was observed only in response to trans-DDP suggesting the involvement of this activity in the downregulation of JNK activity in response to this compound. Altogether the results suggest that the prolonged activation of JNK in response to cis-DDP contributes to cell death induction.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Cisplatino/farmacología , Daño del ADN , Proteínas Quinasas Activadas por Mitógenos , Animales , Línea Celular/efectos de los fármacos , Línea Celular/enzimología , Núcleo Celular/enzimología , Citoplasma/enzimología , Activación Enzimática/efectos de los fármacos , Proteínas Quinasas JNK Activadas por Mitógenos , Ratones , Vanadatos/farmacología
12.
Oncogene ; 14(5): 579-87, 1997 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-9053856

RESUMEN

Conditioned medium from PC12 cells incubated with retinoic acid (RA) increases [3H]thymidine incorporation in normal rat kidney (NRK) fibroblasts and 3D9 epithelial cells. The medium also causes anchorage-independent growth of NRK cells, which is strongly potentiated either in the presence of EGF or after activation of latent forms of transforming growth factors (TGFs) by acidification. These results suggest that RA regulates the release of more than one growth factor by PC12 cells. Conditioned media from control or NGF-treated PC12 cells causes growth of NRK cells in soft agar only after acidification. An increase in expression of the TGF-beta1 gene is coincident with NGF-induced neuronal differentiation of PC12 cells. In addition, RA also causes a dose- and time-dependent increase in content of TGF-beta1 transcripts. This increase is, at least in part, secondary to transcriptional activation. Sequences responsible for the effect of RA and NGF are located in the 5'-flanking region of the TGF-beta1 gene. The TFG-beta1 gene has two promoters and in transient transfection assays RA and NGF significantly enhance the activity of constructs containing the second promoter. High-affinity TGF-beta1 receptors were undetectable in PC12 cells both before and after NGF or RA treatment. RA and NGF decrease PC12 cell proliferation and a neutralizing anti-TGF-beta1 antibody does not reverse this inhibition. In summary, an increase in expression and secretion of TGF-beta1 accompanies RA and NGF-induced PC12 cell growth arrest, but TGF-beta1 does not play an autocrine role in this inhibition.


Asunto(s)
Factores de Crecimiento Nervioso/farmacología , Transcripción Genética , Factor de Crecimiento Transformador beta/biosíntesis , Tretinoina/farmacología , Neoplasias de las Glándulas Suprarrenales , Animales , Diferenciación Celular , División Celular/efectos de los fármacos , Línea Celular , Medios de Cultivo Condicionados , Factor de Crecimiento Epidérmico/farmacología , Epitelio , Fibroblastos , Riñón , Cinética , Ratones , Visón , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/fisiología , Células PC12 , Feocromocitoma , Ratas , Proteínas Recombinantes/biosíntesis , Secuencias Reguladoras de Ácidos Nucleicos , Transcripción Genética/efectos de los fármacos , Transfección , Factor de Crecimiento Transformador beta/genética
13.
Oncogene ; 8(5): 1285-92, 1993 May.
Artículo en Inglés | MEDLINE | ID: mdl-8479750

RESUMEN

rho genes have been found in both lower and higher eucaryotes. They code for proteins of 21 kDa, highly conserved in evolution, which belong to the superfamily of ras GTPases. Among the members of this superfamily there are proteins with a regulatory function, such as ras, and proteins involved in vesicular trafficking, such as the family of rab proteins. We have investigated the putative role of rho proteins from Aplysia californica as transforming GTPases utilizing the wild-type and a Val-14 mutant, equivalent to the oncogenic Val-12 mutation of ras genes found in animal and human tumors. Over-expression of either rho gene was sufficient to confer anchorage- and serum-independent growth. Moreover, when introduced into nude mice, selected clones generated from either gene were able to induce tumors, although those carrying the mutated version were more efficient. Pathological analysis indicated that generated tumors corresponded to well-differentiated fibrosarcomas with distinct and intersecting bundles and spindle cells. By contrast, ras-induced tumors were poorly differentiated fibrosarcomas. Thus, our results indicate that under appropriate conditions rho genes function as oncogenes and may have a role in the regulation of proliferation in fibroblast cells.


Asunto(s)
Aplysia/genética , GTP Fosfohidrolasas/genética , Proteínas de Unión al GTP/genética , Proteínas de la Membrana/genética , Neoplasias Experimentales/etiología , Oncogenes , Células 3T3 , Animales , Secuencia de Bases , Fenómenos Fisiológicos Sanguíneos , División Celular , Transformación Celular Neoplásica , GTP Fosfohidrolasas/fisiología , Proteínas de Unión al GTP/fisiología , Masculino , Proteínas de la Membrana/fisiología , Ratones , Datos de Secuencia Molecular , Transfección , Proteína de Unión al GTP rhoB
14.
Oncogene ; 10(11): 2213-20, 1995 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-7784066

RESUMEN

Signal transduction is the major mechanism by which cells communicate among themselves through extracellular stimuli. Among the different structural components involved in signal transduction, protein kinases are one of the key elements in the process. Protein kinase C is a multimember family of kinases which has been involved in the regulation of diverse cellular functions. Regulation of cell growth in fibroblasts has been reported to be one of such functions. In particular the PKC zeta isoenzyme has been postulated to be transforming to NIH3T3 cells (Berra et al., 1993) and to serve as an effector for Ras proteins through the regulation of the NF kappa B transcription factor (Dominguez et al., 1993) and direct interaction (Díaz-Meco et al., 1994). We have investigated the effects of overexpressing the mouse wild-type PKC zeta in NIH3T3 cells. When compared to the parental NIH3T3 cells, we have found (1) no significant effect on cell morphology; (2) no difference in growth properties in the absence of serum or in the presence of individual growth factors such as insulin, phorbol esters or PDGF; (3) no growth in soft agar nor tumorigenic activity in nude mice. In addition cells stably overexpressing the PKC zeta kinase did not interfere or amplify the induction of NF kappa B activity by tumor necrosis factor alfa (TNF-alpha) nor altered NF kappa B activity in transient expression of cells treated with TNF-alpha. Thus, mammalian PKC zeta is most likely not directly involved in the regulation of cell proliferation in fibroblasts nor affects directly or indirectly the activation of NF kappa B.


Asunto(s)
Transformación Celular Neoplásica , Expresión Génica , FN-kappa B/metabolismo , Proteína Quinasa C/genética , Células 3T3 , Animales , Secuencia de Bases , Sustancias de Crecimiento/metabolismo , Ratones , Datos de Secuencia Molecular , Oligodesoxirribonucleótidos , Transducción de Señal
15.
Oncogene ; 19(45): 5142-52, 2000 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-11064451

RESUMEN

Treatment of cells with cisplatin induces a sustained activation of the stress activated protein kinase SAPK/JNK and the mitogen-activated protein kinase p38. Activation of JNK by cisplatin is necessary for the induction of apoptosis. Expression of the MAPK phosphatases CL100/MKP-1 and hVH-5 selectively prevents JNK/SAPK activation by cisplatin in a dose dependent fashion and results in protection against cisplatin-induced apoptosis. In contrast, expression of the ERK-specific phosphatase Pyst1 inhibits JNK/SAPK activity only when expressed at very high levels and does not confer protection against cisplatin. Furthermore, expression of a catalytically inactive mutant of CL100 in 293 cells decreases the IC50 for cisplatin and increases the toxicity of transplatin. This effect seems to be mediated by an increase in JNK activity since p38 activity is unaffected. These results suggest that dual-specificity MAPK phosphatases may be candidate drug targets in order to optimize cisplatin based therapeutic protocols.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis , Proteínas de Ciclo Celular , Cisplatino/farmacología , Proteínas Inmediatas-Precoces/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosfoproteínas Fosfatasas , Proteínas Tirosina Fosfatasas/metabolismo , Fosfatasa 1 de Especificidad Dual , Fosfatasa 6 de Especificidad Dual , Fosfatasas de Especificidad Dual , Activación Enzimática/efectos de los fármacos , Humanos , Proteínas Inmediatas-Precoces/genética , Proteína Quinasa 8 Activada por Mitógenos , Mutación , Unión Proteica , Proteína Fosfatasa 1 , Proteínas Tirosina Fosfatasas/genética , Estereoisomerismo , Transcripción Genética , Células Tumorales Cultivadas
16.
Oncogene ; 10(5): 811-6, 1995 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-7898922

RESUMEN

Oncogenes appear to influence apoptosis in two ways. Some activate cells from a growth-arrested state to one in which both apoptosis and entry to S-phase become possible, the choice between them being determined by a second signal, such as cytokine or growth factor. Cells in this state are often sensitive to apoptosis induced by a wide variety of agents, including several drugs used in cancer chemotherapy. Other oncogenes prevent activation of the apoptosis effector pathway, even in the presence of a death stimulus, the affected cells therefore being resistant to chemotherapeutic agents. In rodent fibroblasts, c-myc or the adenovirus oncogene E1A effect the first type of change, whereas bcl-2, v-abl, E1B or activated ras effect the second. Here we study in rodent fibroblast the effect of expression of rho genes, members of the ras superfamily which we have previously shown to be tumorigenic when highly expressed in this cell type. We show that expression of wild-type rho from Aplysia californica stimulates apoptosis in cultured cell lines and that the apoptotic index in tumors generated by these cell lines is similar to those induced by E1A-transformed cells. In contrast, mutated rho, activated by Val14 substitution in the GTP binding site, it less potent as a stimulator of apoptosis, generating a phenotype more similar to that obtained with activated ras. Thus, rho genes may play a critical role in the regulation of apoptosis.


Asunto(s)
Apoptosis , Proteínas de Unión al GTP/fisiología , Células 3T3 , Animales , Cisplatino/farmacología , Medio de Cultivo Libre de Suero , ADN/metabolismo , Proteínas de Unión al GTP/genética , Expresión Génica , Ratones , Transfección , Proteínas de Unión al GTP rho
17.
Oncogene ; 17(14): 1855-69, 1998 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-9778052

RESUMEN

Rho proteins are a branch of GTPases that belongs to the Ras superfamily which are critical elements of signal transduction pathways leading to a variety of cellular responses. This family of small GTPases has been involved in diverse biological functions such as cytoskeleton organization, cell growth and transformation, cell motility, migration, metastasis, and responses to stress. We report that several human Rho proteins including Rho A, Rho C and Rac 1, are capable of inducing apoptosis in different cell systems like murine NIH3T3 fibroblasts and the human erythroleukemia K562 cell line. Since K562 cells are devoid of p53, apoptosis induced by Rho in this system is independent of p53. Rho-dependent apoptosis is mediated by the generation of ceramides, and it is drastically inhibited by ectopic expression of Bcl2, both under in vitro and in vivo conditions. Furthermore, the human oncogenes vav and ost that have been shown to function as guanine exchange factors for Rho proteins, were also able to induce apoptosis under similar conditions. Finally, we also report that the levels of endogenous Rho proteins are increased when U937 myeloid leukemia cells are exposed to apoptosis-inducing conditions such as TNF alpha treatment. Furthermore, TNF alpha-induced apoptosis in these cells is inhibited by expression of a dominant negative mutant of Rac 1 but it is not affected by a similar mutant of Rho A. These results suggest that Rho proteins play an important role in the physiological regulation of the apoptotic response to stress-inducing agents.


Asunto(s)
Apoptosis , Proteínas de Unión al GTP/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo , Proteínas de Unión al GTP rho , Células 3T3 , Animales , Ceramidas/biosíntesis , Proteínas de Unión al GTP/genética , Factores de Intercambio de Guanina Nucleótido , Humanos , Ratones , Proteínas/metabolismo , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/farmacología , Proteínas de Unión al GTP rac , Factores de Intercambio de Guanina Nucleótido ras , Proteínas ras , Proteína de Unión al GTP rhoA , Proteína rhoC de Unión a GTP
18.
FEBS Lett ; 453(1-2): 151-8, 1999 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-10403393

RESUMEN

Antineoplasic agents such as cisplatin and adriamycin execute their pharmacological role by triggering apoptosis. We have studied the mechanism of apoptosis induction by cisplatin and adriamycin. Both drugs activated JNK with slow and persistent kinetics. Adriamycin activated caspase-3 before the rise in JNK activity, while the response to cisplatin occurs hours after JNK activation. The increase in JNK activity was necessary for cisplatin-mediated apoptosis but it was dispensable for adriamycin-induced cell death. Cells derived from c-jun knock out mice were more resistant to cisplatin cell death than normal cells, but no difference was observed in response to adriamycin. Activation of JNK and cell death by cisplatin is mediated by the MEKK1/SEK1 cascade, since expression of dominant negative expression vectors of these kinases blocked both processes. p38 was also activated by cisplatin with similar kinetics as JNK. AP-1 complexes were activated by cisplatin including mainly c-jun/ATF-2 heterodimers suggesting that AP-1-dependent transcription partially mediated cisplatin-induced apoptosis.


Asunto(s)
Antineoplásicos/farmacología , Cisplatino/farmacología , MAP Quinasa Quinasa 4 , Quinasa 1 de Quinasa de Quinasa MAP , Quinasas de Proteína Quinasa Activadas por Mitógenos , Proteínas Quinasas Activadas por Mitógenos , Proteínas Proto-Oncogénicas c-jun/deficiencia , Factor de Transcripción Activador 2 , Animales , Apoptosis , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Caspasa 3 , Caspasas/metabolismo , Supervivencia Celular , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/aislamiento & purificación , Doxorrubicina/farmacología , Activación Enzimática , Proteínas Quinasas JNK Activadas por Mitógenos , Ratones , Ratones Noqueados , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-fos/aislamiento & purificación , Proteínas Proto-Oncogénicas c-jun/genética , Transducción de Señal , Factor de Transcripción AP-1/aislamiento & purificación , Factores de Transcripción/aislamiento & purificación
19.
Cell Cycle ; 13(22): 3590-601, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25483095

RESUMEN

Aneuploidy and chromosomal instability (CIN) are common features of gastric cancer (GC), but their contribution to carcinogenesis and antitumour therapy response is still poorly understood. Failures in the mitotic checkpoint induced by changes in expression levels of the spindle assembly checkpoint (SAC) proteins cause the missegregation of chromosomes in mitosis as well as aneuploidy. To evaluate the possible contribution of SAC to GC, we analyzed the expression levels of proteins of the mitotic checkpoint complex in a cohort of GC cell lines. We found that the central SAC proteins, Mad2 and BubR1, were the more prominently expressed members in disseminated GC cell lines. Silencing of Mad2 and BubR1 in MKN45 and ST2957 cells decreased their cell proliferation, migration and invasion abilities, indicating that Mad2 and BubR1 could contribute to cellular transformation and tumor progression in GC. We next evaluated whether silencing of SAC proteins could affect the response to microtubule poisons. We discovered that paclitaxel treatment increased cell survival in MKN45 cells interfered for Mad2 or BubR1 expression. However, apoptosis (assessed by caspase-3 activation, PARP proteolysis and levels of antiapoptotic Bcl 2-family members), the DNA damage response (assessed by H2Ax phosphorylation) and exit from mitosis (assessed by Cyclin B degradation and Cdk1 regulation) were activated equally between cells, independently of Mad2 or BubR1-protein levels. In contrast, we observed that the silencing of Mad2 or BubR1 in MKN45 cells showed the induction of a senescence-like phenotype accompanied by cell enlargement, increased senescence-associated ß-galactosidase activity and increased IL-6 and IL-8 expression. In addition, the senescent phenotype is highly increased after treatment with PTX, indicating that senescence could prevent tumorigenesis in GC. In conclusion, the results presented here suggest that Mad2 and BubR1 could be used as prognostic markers of tumor progression and new pharmacological targets in the treatment for GC.


Asunto(s)
Carcinogénesis , Proteínas Mad2/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Neoplasias Gástricas/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Humanos , Cinetocoros/efectos de los fármacos , Proteínas Mad2/genética , Mitosis/genética , Paclitaxel/uso terapéutico , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal/genética , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/patología
20.
Clin Transl Oncol ; 16(6): 529-38, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24065372

RESUMEN

BACKGROUND: Dyskeratosis congenita (DC) is a rare inherited bone marrow failure syndrome with high clinical heterogeneity. Various mutations have been reported in DC patients, affecting genes that code for components of H/ACA ribonucleoproteins, proteins of the telomerase complex and components of the shelterin complex. OBJECTIVES: We aim to clarify the role of ribosome biogenesis failure in senescence induction in X-DC since some studies in animal models have reported a decrease in ribosome biogenesis as a major role in the disease. METHODS: Dyskerin was depleted in normal human fibroblasts by expressing two DKC1 shRNAs. Common changes in gene expression profile between these dyskerin-depleted cells and X-DC fibroblasts were analyzed. RESULTS: Dyskerin depletion induced early activation of the p53 pathway probably secondary to ribosome biogenesis failure. However, the p53 pathway in the fibroblasts from X-DC patients was activated only after an equivalent number of passes to AD-DC fibroblasts, in which telomere attrition in each division rendered shorter telomeres than control fibroblasts. Indeed, no induction of DNA damage was observed in dyskerin-depleted fibroblasts in contrast to X-DC or AD-DC fibroblasts suggesting that DNA damage induced by telomere attrition is responsible for p53 activation in X-DC and AD-DC fibroblasts. Moreover, p53 depletion in senescent DC fibroblasts rescued their proliferative capacity and reverted the morphological changes produced after prolonged culture. CONCLUSIONS: Our data indicate that ribosome biogenesis do not seem to play an important role in dyskeratosis congenita, conversely increasing DNA damage and activation of p53 pathway triggered by telomere shortening is the main activator of cell senescence.


Asunto(s)
Daño del ADN/genética , Disqueratosis Congénita/genética , Fibroblastos/metabolismo , Ribosomas/fisiología , Telómero/genética , Proteína p53 Supresora de Tumor/genética , Biomarcadores/metabolismo , Western Blotting , Ciclo Celular , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular , Células Cultivadas , Senescencia Celular/fisiología , Disqueratosis Congénita/metabolismo , Disqueratosis Congénita/patología , Fibroblastos/citología , Perfilación de la Expresión Génica , Humanos , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/genética , ARN Interferente Pequeño , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína p53 Supresora de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda