Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Methods Mol Biol ; 2445: 117-125, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34972989

RESUMEN

Chromosomal instability (CIN) is a hallmark of cancer, which is characterized by the gain or loss of chromosomes as well as the rearrangement of the genetic material during cell division. Detection of mitotic errors such as misaligned chromosomes or chromosomal bridges (also known as lagging chromosomes) is challenging as it requires the analysis and manual discrimination of chromosomal aberrations in mitotic cells by molecular techniques. In interphase cells, more frequent in the cell population than mitotic cells, two distinct nuclear phenotypes are associated with CIN: the micronucleus and the toroidal nucleus. Several methods are available for the detection of micronuclei, but none for toroidal nuclei. Here, we provide a method to quantify the presence of both nuclear biomarkers for the evaluation of CIN status in non-mitotic cells particularly suited for genotoxicity screens.


Asunto(s)
Inestabilidad Cromosómica , Mitosis , Biomarcadores , Núcleo Celular/genética , Cromosomas , Humanos , Mitosis/genética
2.
Methods Mol Biol ; 2445: 127-137, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34972990

RESUMEN

The detection of autophagic vesicles in interphase cells is well characterized with markers such as LC3, SQSTM1 (also known as p62) and LAMP2, which are commonly used in immunofluorescence and biochemistry assays to evaluate the status of autophagy in adherent cells. During mitosis, cells undergo important morphological changes which alter the position of the central plane, therefore the imaging of dividing cells has to be specifically designed. Here, we describe a method to label and image autophagic vesicles in mitotic cells to systematically analyze their number, morphology and distribution.


Asunto(s)
Autofagia , Mitosis , Técnica del Anticuerpo Fluorescente , Proteína Sequestosoma-1
3.
Sci Adv ; 7(48): eabg9275, 2021 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-34818049

RESUMEN

Ribosomes execute the transcriptional program in every cell. Critical to sustain nearly all cellular activities, ribosome biogenesis requires the translation of ~200 factors of which 80 are ribosomal proteins (RPs). As ribosome synthesis depends on RP mRNA translation, a priority within the translatome architecture should exist to ensure the preservation of ribosome biogenesis capacity, particularly under adverse growth conditions. Here, we show that under critical metabolic constraints characterized by mTOR inhibition, LARP1 complexed with the 40S subunit protects from ribophagy the mRNAs regulon for ribosome biogenesis and protein synthesis, acutely preparing the translatome to promptly resume ribosomes production after growth conditions return permissive. Characterizing the LARP1-protected translatome revealed a set of 5'TOP transcript isoforms other than RPs involved in energy production and in mitochondrial function, among other processes, indicating that the mTOR-LARP1-5'TOP axis acts at the translational level as a primary guardian of the cellular anabolic capacity.

4.
Autophagy ; 17(3): 796-813, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32573315

RESUMEN

Lysosomes, as primary degradative organelles, are the endpoint of different converging pathways, including macroautophagy. To date, lysosome degradative function has been mainly studied in interphase cells, while their role during mitosis remains controversial. Mitosis dictates the faithful transmission of genetic material among generations, and perturbations of mitotic division lead to chromosomal instability, a hallmark of cancer. Heretofore, correct mitotic progression relies on the orchestrated degradation of mitotic factors, which was mainly attributed to ubiquitin-triggered proteasome-dependent degradation. Here, we show that mitotic transition also relies on lysosome-dependent degradation, as impairment of lysosomes increases mitotic timing and leads to mitotic errors, thus promoting chromosomal instability. Furthermore, we identified several putative lysosomal targets in mitotic cells. Among them, WAPL, a cohesin regulatory protein, emerged as a novel SQSTM1-interacting protein for targeted lysosomal degradation. Finally, we characterized an atypical nuclear phenotype, the toroidal nucleus, as a novel biomarker for genotoxic screenings. Our results establish lysosome-dependent degradation as an essential event to prevent chromosomal instability.Abbreviations: 3D: three-dimensional; APC/C: anaphase-promoting complex; ARL8B: ADP ribosylation factor like GTPase 8B; ATG: autophagy-related; BORC: BLOC-one-related complex; CDK: cyclin-dependent kinase; CENPE: centromere protein E; CIN: chromosomal instability; ConcA: concanamycin A; CQ: chloroquine; DAPI: 4,6-diamidino-2-penylinole; FTI: farnesyltransferase inhibitors; GFP: green fluorescent protein; H2B: histone 2B; KIF: kinesin family member; LAMP2: lysosomal associated membrane protein 2; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MEF: mouse embryonic fibroblast; MTOR: mechanistic target of rapamycin kinase; PDS5B: PDS5 cohesin associated factor B; SAC: spindle assembly checkpoint; PLEKHM2: pleckstrin homology and RUN domain containing M2; SQSTM1: sequestosome 1; TEM: transmission electron microscopy; ULK1: unc-51 like autophagy activating kinase 1; UPS: ubiquitin-proteasome system; v-ATPase: vacuolar-type H+-translocating ATPase; WAPL: WAPL cohesion release factor.


Asunto(s)
Autofagia/fisiología , Inestabilidad Cromosómica/fisiología , Fibroblastos/metabolismo , Lisosomas/metabolismo , Animales , Células HeLa , Humanos , Mitosis/fisiología , Complejo de la Endopetidasa Proteasomal/metabolismo , Factores de Transcripción/metabolismo , Ubiquitina/metabolismo
5.
EMBO J ; 39(13): e103838, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32484960

RESUMEN

Many oncogenes enhance nucleotide usage to increase ribosome content, DNA replication, and cell proliferation, but in parallel trigger p53 activation. Both the impaired ribosome biogenesis checkpoint (IRBC) and the DNA damage response (DDR) have been implicated in p53 activation following nucleotide depletion. However, it is difficult to reconcile the two checkpoints operating together, as the IRBC induces p21-mediated G1 arrest, whereas the DDR requires that cells enter S phase. Gradual inhibition of inosine monophosphate dehydrogenase (IMPDH), an enzyme required for de novo GMP synthesis, reveals a hierarchical organization of these two checkpoints. We find that the IRBC is the primary nucleotide sensor, but increased IMPDH inhibition leads to p21 degradation, compromising IRBC-mediated G1 arrest and allowing S phase entry and DDR activation. Disruption of the IRBC alone is sufficient to elicit the DDR, which is strongly enhanced by IMPDH inhibition, suggesting that the IRBC acts as a barrier against genomic instability.


Asunto(s)
Daño del ADN , Puntos de Control de la Fase G1 del Ciclo Celular , Nucleótidos/metabolismo , Ribosomas/metabolismo , Células HCT116 , Humanos , Nucleótidos/genética , Ribosomas/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
6.
iScience ; 20: 434-448, 2019 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-31627130

RESUMEN

Cancer cells rely on mTORC1 activity to coordinate mitogenic signaling with nutrients availability for growth. Based on the metabolic function of E2F1, we hypothesize that glucose catabolism driven by E2F1 could participate on mTORC1 activation. Here, we demonstrate that glucose potentiates E2F1-induced mTORC1 activation by promoting mTORC1 translocation to lysosomes, a process that occurs independently of AMPK activation. We showed that E2F1 regulates glucose metabolism by increasing aerobic glycolysis and identified the PFKFB3 regulatory enzyme as an E2F1-regulated gene important for mTORC1 activation. Furthermore, PFKFB3 and PFK1 were found associated to lysosomes and we demonstrated that modulation of PFKFB3 activity, either by substrate accessibility or expression, regulates the translocation of mTORC1 to lysosomes by direct interaction with Rag B and subsequent mTORC1 activity. Our results support a model whereby a glycolytic metabolon containing phosphofructokinases transiently interacts with the lysosome acting as a sensor platform for glucose catabolism toward mTORC1 activity.

7.
Cancer Res ; 79(17): 4348-4359, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31292158

RESUMEN

The role of MYC in regulating p53 stability as a function of increased ribosome biogenesis is controversial. On the one hand, it was suggested that MYC drives the overexpression of ribosomal proteins (RP)L5 and RPL11, which bind and inhibit HDM2, stabilizing p53. On the other, it has been proposed that increased ribosome biogenesis leads the consumption of RPL5/RPL11 into nascent ribosomes, reducing p53 levels and enhancing tumorigenesis. Here, we show that the components that make up the recently described impaired ribosome biogenesis checkpoint (IRBC) complex, RPL5, RPL11, and 5S rRNA, are reduced following MYC silencing. This leads to a rapid reduction in p53 protein half-life in an HDM2-dependent manner. In contrast, MYC induction leads to increased ribosome biogenesis and p53 protein stabilization. Unexpectedly, there is no change in free RPL5/RPL11 levels, but there is a striking increase in IRBC complex bound to HDM2. Our data support a cell-intrinsic tumor-suppressor response to MYC expression, which is presently being exploited to treat cancer. SIGNIFICANCE: Oncogenic MYC induces the impaired ribosome biogenesis checkpoint, which could be potentially targeted for cancer treatment.


Asunto(s)
Proteínas Proto-Oncogénicas c-myc/genética , Ribosomas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Línea Celular Tumoral , Proliferación Celular/genética , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Regulación de la Expresión Génica , Humanos , Biosíntesis de Proteínas , Estabilidad Proteica , Proteínas Proto-Oncogénicas c-mdm2/genética , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , ARN Ribosómico 5S/genética , ARN Ribosómico 5S/metabolismo , Proteínas Ribosómicas/genética , Proteínas Ribosómicas/metabolismo , Ribosomas/genética , Proteína p53 Supresora de Tumor/genética
8.
Mol Cell ; 67(1): 55-70.e4, 2017 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-28673543

RESUMEN

Ribosomal protein (RP) expression in higher eukaryotes is regulated translationally through the 5'TOP sequence. This mechanism evolved to more rapidly produce RPs on demand in different tissues. Here we show that 40S ribosomes, in a complex with the mRNA binding protein LARP1, selectively stabilize 5'TOP mRNAs, with disruption of this complex leading to induction of the impaired ribosome biogenesis checkpoint (IRBC) and p53 stabilization. The importance of this mechanism is underscored in 5q− syndrome, a macrocytic anemia caused by a large monoallelic deletion, which we found to also encompass the LARP1 gene. Critically, depletion of LARP1 alone in human adult CD34+ bone marrow precursor cells leads to a reduction in 5'TOP mRNAs and the induction of p53. These studies identify a 40S ribosome function independent of those in translation that, with LARP1, mediates the autogenous control of 5'TOP mRNA stability, whose disruption is implicated in the pathophysiology of 5q− syndrome.


Asunto(s)
Autoantígenos/metabolismo , Biosíntesis de Proteínas , Secuencia de Oligopirimidina en la Región 5' Terminal del ARN , Estabilidad del ARN , ARN Mensajero/metabolismo , Ribonucleoproteínas/metabolismo , Proteínas Ribosómicas/metabolismo , Ribosomas/metabolismo , Anemia Macrocítica/genética , Anemia Macrocítica/metabolismo , Autoantígenos/genética , Células de la Médula Ósea/metabolismo , Deleción Cromosómica , Cromosomas Humanos Par 5/genética , Cromosomas Humanos Par 5/metabolismo , Células HCT116 , Humanos , Complejos Multiproteicos , Unión Proteica , Interferencia de ARN , ARN Mensajero/genética , Ribonucleoproteínas/genética , Proteínas Ribosómicas/genética , Ribosomas/genética , Factores de Tiempo , Transfección , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Antígeno SS-B
9.
Mol Cancer ; 15: 1, 2016 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-26728659

RESUMEN

BACKGROUND: Neuroblastoma is a malignant embryonal tumor occurring in young children, consisting of undifferentiated neuroectodermal cells derived from the neural crest. Current therapies for high-risk neuroblastoma are insufficient, resulting in high mortality rates and high incidence of relapse. With the intent to find new therapies for neuroblastomas, we investigated the efficacy of low-doses of actinomycin D, which at low concentrations preferentially inhibit RNA polymerase I-dependent rRNA trasncription and therefore, ribosome biogenesis. METHODS: Neuroblastoma cell lines with different p53 genetic background were employed to determine the response on cell viability and apoptosis of low-dose of actinomycin D. Subcutaneously-implanted SK-N-JD derived neuroblastoma tumors were used to assess the effect of low-doses of actinomycin D on tumor formation. RESULTS: Low-dose actinomycin D treatment causes a reduction of cell viability in neuroblastoma cell lines and that this effect is stronger in cells that are wild-type for p53. MYCN overexpression contributes to enhance this effect, confirming the importance of this oncogene in ribosome biogenesis. In the wild-type SK-N-JD cell line, apoptosis was the major mechanism responsible for the reduction in viability and we demonstrate that treatment with the MDM2 inhibitor Nutlin-3, had a similar effect to that of actinomycin D. Apoptosis was also detected in p53(-/-)deficient LA1-55n cells treated with actinomycin D, however, only a small recovery of cell viability was found when apoptosis was inhibited by a pan-caspase inhibitor, suggesting that the treatment could activate an apoptosis-independent cell death pathway in these cells. We also determined whether actinomycin D could increase the efficacy of the histone deacetylase inhibitor, SAHA, which is in being used in neuroblastoma clinical trials. We show that actinomycin D synergizes with SAHA in neuroblastoma cell lines. Moreover, on subcutaneously-implanted neuroblastoma tumors derived from SK-N-JD cells, actinomycin D led to tumor regression, an effect enhanced in combination with SAHA. CONCLUSIONS: The results presented in this work demonstrate that actinomycin D, at low concentrations, inhibits proliferation and induces cell death in vitro, as well as tumor regression in vivo. From this study, we propose that use of ribosome biogenesis inhibitors should be clinically considered as a potential therapy to treat neuroblastomas.


Asunto(s)
Dactinomicina/uso terapéutico , Neuroblastoma/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Dactinomicina/farmacología , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Femenino , Ácidos Hidroxámicos/farmacología , Imidazoles/farmacología , Ratones , Neuroblastoma/patología , Piperazinas/farmacología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Factores de Tiempo , Proteína p53 Supresora de Tumor/metabolismo , Vorinostat
10.
Oncotarget ; 6(29): 28057-70, 2015 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-26356814

RESUMEN

In addition to being a master regulator of cell cycle progression, E2F1 regulates other associated biological processes, including growth and malignancy. Here, we uncover a regulatory network linking E2F1 to lysosomal trafficking and mTORC1 signaling that involves v-ATPase regulation. By immunofluorescence and time-lapse microscopy we found that E2F1 induces the movement of lysosomes to the cell periphery, and that this process is essential for E2F1-induced mTORC1 activation and repression of autophagy. Gain- and loss-of-function experiments reveal that E2F1 regulates v-ATPase activity and inhibition of v-ATPase activity repressed E2F1-induced lysosomal trafficking and mTORC1 activation. Immunoprecipitation experiments demonstrate that E2F1 induces the recruitment of v-ATPase to lysosomal RagB GTPase, suggesting that E2F1 regulates v-ATPase activity by enhancing the association of V0 and V1 v-ATPase complex. Analysis of v-ATPase subunit expression identified B subunit of V0 complex, ATP6V0B, as a transcriptional target of E2F1. Importantly, ATP6V0B ectopic-expression increased v-ATPase and mTORC1 activity, consistent with ATP6V0B being responsible for mediating the effects of E2F1 on both responses. Our findings on lysosomal trafficking, mTORC1 activation and autophagy suppression suggest that pharmacological intervention at the level of v-ATPase may be an efficacious avenue for the treatment of metastatic processes in tumors overexpressing E2F1.


Asunto(s)
Factor de Transcripción E2F1/metabolismo , Complejos Multiproteicos/metabolismo , Neoplasias/patología , Transporte de Proteínas/fisiología , Serina-Treonina Quinasas TOR/metabolismo , ATPasas de Translocación de Protón Vacuolares/metabolismo , Autofagia/fisiología , Western Blotting , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Técnica del Anticuerpo Fluorescente , Humanos , Inmunoprecipitación , Lisosomas/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina , ARN Interferente Pequeño , Reacción en Cadena en Tiempo Real de la Polimerasa , Transfección
11.
Cell Oncol (Dordr) ; 38(5): 341-52, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26306783

RESUMEN

BACKGROUND: In the past, the HDAC inhibitor suberoylanilide hydroxamic acid (SAHA) has been shown to induce apoptosis in several human tumor types, including neuroblastomas. Amplification and over-expression of the MYCN oncogene is a diagnostic hallmark and a poor prognostic indicator in high-risk neuroblastomas. Here, we studied the relationship between MYCN amplification and over-expression and the anti-tumor effect of SAHA to assess whether this drug may serve as a treatment option for high-risk neuroblastomas. METHODS: Different human neuroblastoma cell lines, over-expressing or not over-expressing MYCN, were used in this study. Targeted knockdown and exogenous over-expression of MYCN were employed to examine correlations between MYCN expression levels and SAHA responses. After various time periods and concentration exposures to the drug, cell viability was measured by MTS assay, and variations in MYCN mRNA and protein levels were assessed by qPCR and Western blotting, respectively. RESULTS: We found that SAHA decreased cell viability in all cell lines tested through apoptosis induction, and that SAHA had a stronger effect on cell lines carrying an amplified MYCN gene. A decrease in MYCN mRNA and protein levels was observed in the SAHA treated cell lines. Subsequent silencing and exogenous over-expression of MYCN changed the proliferation rate of the cells, but did not have any significant impact on the effect of SAHA on the viability of the cells. We also found that SAHA blocked the expression of MYCN and, by doing so, reduced the effects mediated by this protein. CONCLUSIONS: Our results suggest that SAHA may be used as a single-drug treatment option for neuroblastomas with an amplified MYCN gene, and as an adjuvant treatment option for all neuroblastomas.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Ácidos Hidroxámicos/farmacología , Proteínas Nucleares/genética , Proteínas Oncogénicas/genética , Apoptosis/efectos de los fármacos , Apoptosis/genética , Western Blotting , Línea Celular Tumoral , Proliferación Celular/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Relación Dosis-Respuesta a Droga , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Microscopía Fluorescente , Proteína Proto-Oncogénica N-Myc , Neuroblastoma/genética , Neuroblastoma/metabolismo , Neuroblastoma/patología , Proteínas Nucleares/metabolismo , Proteínas Oncogénicas/metabolismo , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Vorinostat
12.
J Clin Invest ; 125(7): 2736-47, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-26075820

RESUMEN

Type 2 diabetes mellitus (T2DM) is a worldwide heath problem that is characterized by insulin resistance and the eventual loss of ß cell function. As recent studies have shown that loss of ribosomal protein (RP) S6 kinase 1 (S6K1) increases systemic insulin sensitivity, S6K1 inhibitors are being pursued as potential agents for improving insulin resistance. Here we found that S6K1 deficiency in mice also leads to decreased ß cell growth, intrauterine growth restriction (IUGR), and impaired placental development. IUGR is a common complication of human pregnancy that limits the supply of oxygen and nutrients to the developing fetus, leading to diminished embryonic ß cell growth and the onset of T2DM later in life. However, restoration of placental development and the rescue of IUGR by tetraploid embryo complementation did not restore ß cell size or insulin levels in S6K1-/- embryos, suggesting that loss of S6K1 leads to an intrinsic ß cell lesion. Consistent with this hypothesis, reexpression of S6K1 in ß cells of S6K1-/- mice restored embryonic ß cell size, insulin levels, glucose tolerance, and RPS6 phosphorylation, without rescuing IUGR. Together, these data suggest that a nutrient-mediated reduction in intrinsic ß cell S6K1 signaling, rather than IUGR, during fetal development may underlie reduced ß cell growth and eventual development of T2DM later in life.


Asunto(s)
Retardo del Crecimiento Fetal/enzimología , Retardo del Crecimiento Fetal/patología , Células Secretoras de Insulina/enzimología , Células Secretoras de Insulina/patología , Proteínas Quinasas S6 Ribosómicas 90-kDa/fisiología , Animales , Tamaño de la Célula , Diabetes Mellitus Tipo 2/enzimología , Diabetes Mellitus Tipo 2/etiología , Diabetes Mellitus Tipo 2/patología , Femenino , Prueba de Complementación Genética , Humanos , Insulina/metabolismo , Resistencia a la Insulina , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Placentación/genética , Placentación/fisiología , Embarazo , Embarazo en Diabéticas/enzimología , Embarazo en Diabéticas/patología , Proteínas Quinasas S6 Ribosómicas 90-kDa/deficiencia , Proteínas Quinasas S6 Ribosómicas 90-kDa/genética , Tetraploidía
13.
PLoS One ; 7(12): e51544, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23251571

RESUMEN

In this study we demonstrate that accumulation of reactive oxygen species (ROS) is essential for E2F1 mediated apoptosis in ER-E2F1 PC12 pheochromocytoma, and SH-SY5Y and SK-N-JD neuroblastoma stable cell lines. In these cells, the ER-E2F1 fusion protein is expressed in the cytosol; the addition of 4-hydroxytamoxifen (OHT) induces its translocation to the nucleus and activation of E2F1target genes. Previously we demonstrated that, in ER-E2F1 PC12 cells, OHT treatment induced apoptosis through activation of caspase-3. Here we show that caspase-8 activity did not change upon treatment with OHT. Moreover, over-expression of Bcl-xL arrested OHT-induced apoptosis; by contrast, over-expression of c-FLIP, did not have any effect on OHT-induced apoptosis. OHT addition induces BimL expression, its translocation to mitochondria and activation of Bax, which is paralleled by diminished mitochondrial enrichment of Bcl-xL. Treatment with a Bax-inhibitory peptide reduced OHT-induced apoptosis. These results point out the essential role of mitochondria on the apoptotic process driven by E2F1. ROS accumulation followed E2F1 induction and treatment with the antioxidant N-acetylcysteine, inhibited E2F1-induced Bax translocation to mitochondria and subsequent apoptosis. The role of ROS in mediating OHT-induced apoptosis was also studied in two neuroblastoma cell lines, SH-SY5Y and SK-N-JD. In SH-SY5Y cells, activation of E2F1 by the addition of OHT induced ROS production and apoptosis, whereas over-expression of E2F1 in SK-N-JD cells failed to induce either response. Transcriptional profiling revealed that many of the genes responsible for scavenging ROS were down-regulated following E2F1-induction in SH-SY5Y, but not in SK-N-JD cells. Finally, inhibition of GSK3ß blocked ROS production, Bax activation and the down regulation of ROS scavenging genes. These findings provide an explanation for the apparent contradictory role of E2F1 as an apoptotic agent versus a cell cycle activator.


Asunto(s)
Apoptosis , Factor de Transcripción E2F1/metabolismo , Neuroblastoma/metabolismo , Neuroblastoma/patología , Feocromocitoma/metabolismo , Feocromocitoma/patología , Especies Reactivas de Oxígeno/metabolismo , Neoplasias de las Glándulas Suprarrenales/metabolismo , Neoplasias de las Glándulas Suprarrenales/patología , Animales , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Línea Celular Tumoral , Humanos , Mitocondrias/metabolismo , Multimerización de Proteína , Transporte de Proteínas , Ratas , Transducción de Señal , Proteína X Asociada a bcl-2/metabolismo , Proteína bcl-X/metabolismo
14.
Cancer Lett ; 318(1): 42-52, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22186300

RESUMEN

Neuroblastoma (NB) is the most common solid extracranial tumor in children. Here we showed that trichostatin A, a histone deacetylase inhibitor (HDACi), decreases cell viability in three NB cell lines of different phenotypes. The treatment leads to G2/M-phase arrest, apoptosis and autophagy. Autophagy induction accompanies apoptosis in the most proliferative, N-Myc overexpressing cells. In contrast, autophagy precedes apoptosis and acts as a protective mechanism in the less proliferative, non-N-Myc overexpressing cells. Therefore, the autophagy induction is a relevant event in the NB response to HDACis, and it should be considered in the design of new treatments for this malignancy.


Asunto(s)
Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Ácidos Hidroxámicos/farmacología , Neuroblastoma/patología , Acetilación/efectos de los fármacos , Western Blotting , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Histona Desacetilasas/química , Histona Desacetilasas/metabolismo , Humanos , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa
15.
PLoS One ; 6(1): e16163, 2011 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-21283628

RESUMEN

During cell proliferation, growth must occur to maintain homeostatic cell size. Here we show that E2F1 is capable of inducing growth by regulating mTORC1 activity. The activation of cell growth and mTORC1 by E2F1 is dependent on both E2F1's ability to bind DNA and to regulate gene transcription, demonstrating that a gene induction expression program is required in this process. Unlike E2F1, E2F3 is unable to activate mTORC1, suggesting that growth activity could be restricted to individual E2F members. The effect of E2F1 on the activation of mTORC1 does not depend on Akt. Furthermore, over-expression of TSC2 does not interfere with the effect of E2F1, indicating that the E2F1-induced signal pathway can compensate for the inhibitory effect of TSC2 on Rheb. Immunolocalization studies demonstrate that E2F1 induces the translocation of mTORC1 to the late endosome vesicles, in a mechanism dependent of leucine. E2F1 and leucine, or insulin, together affect the activation of S6K stronger than alone suggesting that they are complementary in activating the signal pathway. From these studies, E2F1 emerges as a key protein that integrates cell division and growth, both of which are essential for cell proliferation.


Asunto(s)
Procesos de Crecimiento Celular , Factor de Transcripción E2F1/fisiología , Proteínas/metabolismo , Transducción de Señal/fisiología , Ciclo Celular , División Celular , Línea Celular Tumoral , Proliferación Celular , Humanos , Insulina/farmacología , Leucina/farmacología , Diana Mecanicista del Complejo 1 de la Rapamicina , Complejos Multiproteicos , Transporte de Proteínas , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Serina-Treonina Quinasas TOR , Activación Transcripcional
16.
Biochim Biophys Acta ; 1803(8): 912-8, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20447425

RESUMEN

Multiple E2F1 phosphorylation sites have been described as targets of different kinases, yet their in vivo implication is uncertain. We previously reported that GSK3beta is able to phosphorylate E2F1 in vitro at Ser403 and Ser433. Recently, it has been shown that both residues are also direct targets of p38 MAP kinase. In order to determine whether Ser403 phosphorylation occurs in vivo and to elucidate its role in E2F1 transcription activity, we developed a phospho-E2F1(Ser403) antibody for use in in vivo detection studies. Our results demonstrate that endogenous E2F1 is phosphorylated in vivo on Ser403, however neither GSK3beta nor p38 MAP kinase are responsible for this event. E2F1 phosphorylation on Ser403 is induced after treatment with doxorubicin in a dose response manner. The transcriptional response of E2F1 to doxorubicin is lower in an E2F1 Ser/Ala403 mutated construct relative to the wild type, suggesting a role for Ser403 phosphorylation in DNA damage conditions. Comparative study between the expression of the bcl2 gene family induced by the wild type and E2F1 Ser/Ala403 mutant revealed a statistically different pattern between both conditions. These results suggest that phosphorylation of Ser403 could influence the selection and regulation of E2F1 target genes.


Asunto(s)
Anticuerpos Fosfo-Específicos/metabolismo , Factor de Transcripción E2F1/metabolismo , Serina/metabolismo , Animales , Antibióticos Antineoplásicos/farmacología , Línea Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Doxorrubicina/farmacología , Factor de Transcripción E2F1/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Fosforilación , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
17.
Biochim Biophys Acta ; 1783(1): 144-52, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17900713

RESUMEN

Mitogenic response to growth factors is concomitant with the modulation they exert on the levels of Fructose 2,6-bisphosphate (Fru-2,6-P2), an essential activator of the glycolytic flux. In mammalian cells, decreased Fru-2,6-P2 concentration causes cell cycle delay, whereas high levels of Fru-2,6-P2 sensitize cells to apoptosis. In order to analyze the cell cycle consequences due to changes in Fru-2,6-P2 levels, the bisphosphatase-dead mutant (H258A) of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase enzyme was over-expressed in Schizosaccharomyces pombe cells and the variation in cell phenotype was studied. The results obtained demonstrate that the increase in Fru-2,6-P2 levels results in a defective division of S. pombe, as revealed by an altered multisepted phenotype. The H258A-expressing cells showed impairment of cytokinesis, but normal nuclear division. In order to identify cellular mediators responsible for this effect, we transformed different S. pombe strains and observed that the cytokinetic defect was absent in cells defective for Wee1 kinase function. Therefore, in S. pombe, Wee1 integrates the metabolic signal emerging from changes in Fru-2,6-P2 content, thus coupling metabolism with cell proliferation. As the key regulators of the cell cycle checkpoints are conserved throughout evolution, these results may help to understand the experimental evidences obtained by manipulation of Fru-2,6-P2 levels in mammalian cells.


Asunto(s)
Fructosadifosfatos/metabolismo , Schizosaccharomyces/citología , Schizosaccharomyces/metabolismo , División Celular , Regulación Fúngica de la Expresión Génica , Mutación/genética , Fosfofructoquinasa-2/genética , Fosfofructoquinasa-2/metabolismo , Schizosaccharomyces/genética , Proteínas de Schizosaccharomyces pombe/genética , Proteínas de Schizosaccharomyces pombe/metabolismo , Transactivadores/genética , Transactivadores/metabolismo
18.
J Neurochem ; 102(6): 2020-2028, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17555552

RESUMEN

Both E2F1 and GSK3beta have been described as essential targets in neuronal apoptosis. Previous studies have demonstrated that GSK3beta binds to E2F1 in vivo. We wanted to investigate whether these proteins could share a common apoptotic signal pathway in neuronal cells. With this intention, we developed a PC12 ER-E2F1 stable cell line in which E2F1 activity was dependent on the presence of 4-hydroxitamoxifen. E2F1 activation produced apoptosis in naive and post-mitotic cells; serum and nerve growth factor respectively protected them from E2F1 apoptotic stimuli. The presence of specific GSK3beta inhibitors SB216763 and LiCl completely protected cells from apoptosis induced by E2F1 activation. In addition, knocked down GSK3beta experiments by small interference RNAs have demonstrated that a reduction of GSK3beta protein levels can lower the apoptotic effect of E2F1. Finally, we demonstrated that the apoptotic effect of E2F1 is not due to the regulation of GSK3beta activity, and that the inhibitory effect of GSK3beta inhibitor SB216763 on E2F1 induced apoptosis could be due to an alteration in the E2F1-regulated transcription gene pattern. In summary, we have demonstrated that the apoptotic action of E2F1 requires GSK3beta activity.


Asunto(s)
Apoptosis/fisiología , Factor de Transcripción E2F1/fisiología , Glucógeno Sintasa Quinasa 3/metabolismo , Neuronas/metabolismo , Transducción de Señal/fisiología , Animales , Apoptosis/efectos de los fármacos , Regulación hacia Abajo/genética , Inhibidores Enzimáticos/farmacología , Antagonistas de Estrógenos/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/fisiología , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3 beta , Neuronas/efectos de los fármacos , Células PC12 , ARN Interferente Pequeño/genética , Ratas , Transducción de Señal/efectos de los fármacos , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacología , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/genética
19.
Biochim Biophys Acta ; 1773(3): 375-82, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17050006

RESUMEN

GSK3beta and E2F1 play an important role in the control of proliferation and apoptosis. Previous work has demonstrated that GSK3beta indirectly regulates E2F activity through modulation of cyclin D1 levels. In this work we show that GSK3beta phosphorylates human E2F1 in vitro at serine 403 and threonine 433, both residues localized at its transactivation domain. This phosphorylation was not detected in vivo. However, co-immunoprecipitation experiments do reveal in vivo binding of these proteins. Moreover, uninhibitable and catalitycally inactive GSK3beta forms inhibit the transcriptional activity of a fusion protein containing E2F1 transactivation domain. Both forms of GSK3beta inhibit E2F1 with similar efficiency. Interestingly the effect was independent of the mutation of serine 403 and threonine 433 to alanine. This suggests that this transcriptional modulation is independent of GSK3beta kinase activity and phosphorylation state of serine 403 and threonine 433. The re-targeting of these GSK3beta forms to the nucleus results in a higher capacity to regulate E2F1 transcriptional activity. Depletion of the levels of GSK3beta protein using siRNA activates E2F1 transcriptional activity. The data presented in this study offer a new mechanism of regulation of E2F1 by direct binding of GSK3beta to its transactivation domain.


Asunto(s)
Factor de Transcripción E2F1/metabolismo , Regulación Enzimológica de la Expresión Génica , Glucógeno Sintasa Quinasa 3/metabolismo , Transactivadores/metabolismo , Línea Celular , Factor de Transcripción E2F1/genética , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3 beta , Humanos , Fosfoserina/metabolismo , Fosfotreonina/metabolismo , Unión Proteica , ARN Interferente Pequeño/genética , Transactivadores/genética
20.
Biochem J ; 368(Pt 1): 283-91, 2002 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-12139485

RESUMEN

In the present study, we demonstrate that E2F is implicated in the regulation of the glycolytic enzyme 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase (6PF2K/Fru-2,6-BPase) during cell division. The expression of this enzyme is induced during the G(1)/S transition of the cell cycle. We identified and monitored the E2F-pocket protein complexes that bind to the E2F site of the F-type promoter during cell-cycle entry, and we analysed their contribution to the phosphatidylinositol 3-kinase (PI 3-kinase)-mediated regulation of the promoter. We found that the predominant E2F complex bound to the F-type promoter in unstimulated/quiescent cells contains E2F4, DP1 and p130 proteins. In serum-stimulated (S-phase) cells, the composition of the complex switched to E2F1/4, DP1 and p107, together with cyclin A and cyclin-dependent kinase 2. Treatment with the PI 3-kinase specific inhibitor LY 294002 prevented the formation of the S-phase complex, suggesting that activation of the PI 3-kinase pathway is essential for the formation of this complex. Further supporting this idea, we obtained results showing that treatment of cycling NIH 3T3 cells with either wortmannin or LY 294002 induces the accumulation of the transcriptionally repressive p130-E2F4-DP1 complex. Using the Rat-1 ER-E2F1 cell line where E2F1 activity can be conditionally induced, we demonstrated that E2F activity is involved in the in vivo transcriptional regulation of the F-type 6PF2K/Fru-2, 6-BPase gene. Taken together, our results show that the F-type 6PF2K/Fru-2, 6-BPase is a genuine E2F-regulated gene, and that its regulation by the PI 3-kinase pathway is at least partially mediated through the E2F transcription factor.


Asunto(s)
Proteínas de Ciclo Celular , Proteínas de Unión al ADN , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfofructoquinasa-2/metabolismo , Factores de Transcripción/metabolismo , Células 3T3 , Animales , Anticuerpos , Sitios de Unión , Células Cultivadas , Cromonas/farmacología , ADN/metabolismo , Factores de Transcripción E2F , Factor de Transcripción E2F1 , Factor de Transcripción E2F4 , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Ratones , Morfolinas/farmacología , Fosfofructoquinasa-2/genética , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...