Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Oncogene ; 36(37): 5296-5308, 2017 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-28504713

RESUMEN

The proto-oncogene PTTG and its binding partner PBF have been widely studied in multiple cancer types, particularly thyroid and colorectal, but their combined role in tumourigenesis is uncharacterised. Here, we show for the first time that together PTTG and PBF significantly modulate DNA damage response (DDR) genes, including p53 target genes, required to maintain genomic integrity in thyroid cells. Critically, DDR genes were extensively repressed in primary thyrocytes from a bitransgenic murine model (Bi-Tg) of thyroid-specific PBF and PTTG overexpression. Irradiation exposure to amplify p53 levels further induced significant repression of DDR genes in Bi-Tg thyrocytes (P=2.4 × 10-4) compared with either PBF- (P=1.5 × 10-3) or PTTG-expressing thyrocytes (P=NS). Consistent with this, genetic instability was greatest in Bi-Tg thyrocytes with a mean genetic instability (GI) index of 35.8±2.6%, as well as significant induction of gross chromosomal aberrations in thyroidal TPC-1 cells following overexpression of PBF and PTTG. We extended our findings to human thyroid cancer using TCGA data sets (n=322) and found striking correlations with PBF and PTTG expression in well-characterised DDR gene panel RNA-seq data. In addition, genetic associations and transient transfection identified PBF as a downstream target of the receptor tyrosine kinase-BRAF signalling pathway, emphasising a role for PBF as a novel component in a pathway well described to drive neoplastic growth. We also showed that overall survival (P=1.91 × 10-5) and disease-free survival (P=4.9 × 10-5) was poorer for TCGA patients with elevated tumoural PBF/PTTG expression and mutationally activated BRAF. Together our findings indicate that PBF and PTTG have a critical role in promoting thyroid cancer that is predictive of poorer patient outcome.


Asunto(s)
Daño del ADN , Proteínas de la Membrana/metabolismo , Securina/metabolismo , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Péptidos y Proteínas de Señalización Intracelular , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Transgénicos , Pronóstico , Proto-Oncogenes Mas , Securina/genética , Tasa de Supervivencia , Neoplasias de la Tiroides/patología , Transfección , Resultado del Tratamiento
2.
J Virol ; 87(18): 10348-55, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23864635

RESUMEN

Human adenovirus E1A makes extensive connections with the cellular protein interaction network. By doing so, E1A can manipulate many cellular programs, including cell cycle progression. Through these reprogramming events, E1A functions as a growth-promoting oncogene and has been used extensively to investigate mechanisms contributing to oncogenesis. Nevertheless, it remains unclear how the C-terminal region of E1A contributes to oncogenic transformation. Although this region is required for transformation in cooperation with E1B, it paradoxically suppresses transformation in cooperation with activated Ras. Previous analysis has suggested that the interaction of E1A with CtBP plays a pivotal role in both activities. However, some C-terminal mutants of E1A retain CtBP binding and yet exhibit defects in transformation, suggesting that other targets of this region are also necessary. To explore the roles of these additional factors, we performed an extensive mutational analysis of the C terminus of E1A. We identified key residues that are specifically required for binding all known targets of the C terminus of E1A. We further tested each mutant for the ability to both localize to the nucleus and transform primary rat cells in cooperation with E1B-55K or Ras. Interaction of E1A with importin α3/Qip1, dual-specificity tyrosine-regulated kinase 1A (DYRK1A), HAN11, and CtBP influenced transformation with E1B-55K. Interestingly, the interaction of E1A with DYRK1A and HAN11 appeared to play a role in suppression of transformation by activated Ras whereas interaction with CtBP was not necessary. This unexpected result suggests a need for revision of current models and provides new insight into transformation by the C terminus of E1A.


Asunto(s)
Adenoviridae/patogenicidad , Proteínas E1A de Adenovirus/metabolismo , Oxidorreductasas de Alcohol/metabolismo , Transformación Celular Viral , Proteínas de Unión al ADN/metabolismo , Adenoviridae/genética , Proteínas E1A de Adenovirus/genética , Animales , Línea Celular , Análisis Mutacional de ADN , Humanos , Mapeo de Interacción de Proteínas , Ratas
3.
Oncogene ; 32(39): 4622-33, 2013 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-23160376

RESUMEN

The anaphase-promoting complex/cyclosome (APC/C) is an ubiquitin ligase that functions during mitosis. Here we identify the transcriptional regulator, transcriptional intermediary factor 1γ, TIF1γ, as an APC/C-interacting protein that regulates APC/C function. TIF1γ is not a substrate for APC/C-dependent ubiquitylation but instead, associates specifically with the APC/C holoenzyme and Cdc20 to affect APC/C activity and progression through mitosis. RNA interference studies indicate that TIF1γ knockdown results in a specific reduction in APC/C ubiquitin ligase activity, the stabilization of APC/C substrates, and an increase in the time taken for cells to progress through mitosis from nuclear envelope breakdown to anaphase. TIF1γ knockdown cells are also characterized by the inappropriate presence of cyclin A at metaphase, and an increase in the number of cells that fail to undergo metaphase-to-anaphase transition. Expression of a small interfering RNA-resistant TIF1γ species relieves the mitotic phenotype imposed by TIF1γ knockdown and allows for mitotic progression. Binding studies indicate that TIF1γ is also a component of the APC/C-mitotic checkpoint complex (MCC), but is not required for MCC dissociation from the APC/C once the spindle assembly checkpoint (SAC) is satisfied. TIF1γ inactivation also results in chromosome misalignment at metaphase and SAC activation; inactivation of the SAC relieves the mitotic block imposed by TIF1γ knockdown. Together these data define novel functions for TIF1γ during mitosis and suggest that a reduction in APC/C ubiquitin ligase activity promotes SAC activation.


Asunto(s)
Mitosis/fisiología , Factores de Transcripción/fisiología , Complejos de Ubiquitina-Proteína Ligasa/metabolismo , Anafase , Ciclosoma-Complejo Promotor de la Anafase , Subunidad Apc3 del Ciclosoma-Complejo Promotor de la Anafase , Proteínas Cdc20 , Proteínas de Ciclo Celular/metabolismo , Células HeLa , Humanos , Ligandos , Puntos de Control de la Fase M del Ciclo Celular , Espectrometría de Masas , Microscopía por Video , Proteínas de Neoplasias/metabolismo , Mapeo de Interacción de Proteínas , Procesamiento Proteico-Postraduccional/fisiología , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Proteínas Recombinantes de Fusión/fisiología , Securina , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/química , Factores de Transcripción/genética , Complejos de Ubiquitina-Proteína Ligasa/fisiología , Ubiquitina-Proteína Ligasas/fisiología , Ubiquitinación/fisiología
4.
Endocrinology ; 153(7): 3526-36, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22535767

RESUMEN

Within the basolateral membrane of thyroid follicular epithelial cells, two transporter proteins are central to thyroid hormone (TH) biosynthesis and secretion. The sodium iodide symporter (NIS) delivers iodide from the bloodstream into the thyroid, and after TH biosynthesis, monocarboxylate transporter 8 (MCT8) mediates TH secretion from the thyroid gland. Pituitary tumor-transforming gene-binding factor (PBF; PTTG1IP) is a protooncogene that is up-regulated in thyroid cancer and that binds NIS and modulates its subcellular localization and function. We now show that PBF binds MCT8 in vitro, eliciting a marked shift in MCT8 subcellular localization and resulting in a significant reduction in the amount of MCT8 at the plasma membrane as determined by cell surface biotinylation assays. Colocalization and interaction between PBF and Mct8 was also observed in vivo in a mouse model of thyroid-specific PBF overexpression driven by a bovine thyroglobulin (Tg) promoter (PBF-Tg). Thyroidal Mct8 mRNA and protein expression levels were similar to wild-type mice. Critically, however, PBF-Tg mice demonstrated significantly enhanced thyroidal TH accumulation and reduced TH secretion upon TSH stimulation. Importantly, Mct8-knockout mice share this phenotype. These data show that PBF binds and alters the subcellular localization of MCT8 in vitro, with PBF overexpression leading to an accumulation of TH within the thyroid in vivo. Overall, these studies identify PBF as the first protein to interact with the critical TH transporter MCT8 and modulate its function in vivo. Furthermore, alongside NIS repression, PBF may thus represent a new regulator of TH biosynthesis and secretion.


Asunto(s)
Proteínas Portadoras/metabolismo , Regulación de la Expresión Génica , Hormonas Tiroideas/metabolismo , Animales , Biotinilación , Células COS , Chlorocebus aethiops , ADN Complementario/metabolismo , Glutatión Transferasa/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Proteínas de Transporte de Membrana/metabolismo , Ratones , Modelos Biológicos , Transportadores de Ácidos Monocarboxílicos , Fenotipo , Procesamiento Proteico-Postraduccional , Simportadores , Tetraspanina 30/biosíntesis , Transcripción Genética
5.
Oncogene ; 28(5): 686-97, 2009 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-19029952

RESUMEN

Using mass spectrometric analysis insulin receptor substrate 4 (IRS-4) has been identified as a novel adenovirus 5 early region 1A (Ad5E1A)-binding protein. IRS-4 interacts with both the transcriptional activation domain (conserved region 3) and the N-terminal region of Ad5E1A13S. Prolonged expression of Ad5E1A13S is required for the observed dramatic increase in the levels of IRS-4 mRNA and protein in Ad5E1-transformed human cell lines. Once expressed, as well as binding to E1A and the insulin receptor, IRS-4 remains tyrosine phosphorylated and constitutively associates with the regulatory p85 subunit of phosphoinositide 3 kinase, resulting in the phosphorylation of Akt (causing activation) and GSK-3beta (causing inhibition). Reducing IRS-4 expression using small interfering RNA (siRNA) in established Ad5E1A-expressing cell lines decreases the activation of Akt and cellular proliferation. During Ad5 infection, IRS-4 is not expressed. However, Ad5E1A associates with IRS-1, increasing Akt and GSK-3beta phosphorylation and tyrosine phosphorylation of IRS-1 itself. We conclude that the association and altered regulation of IRS proteins by Ad5E1A contribute to the adenovirus-transformed phenotype and modulates viral infection in an Akt-dependent manner.


Asunto(s)
Proteínas E1A de Adenovirus/metabolismo , Proteínas E1A de Adenovirus/fisiología , Transformación Celular Viral , Proteínas Sustrato del Receptor de Insulina/metabolismo , Proteína Oncogénica v-akt/fisiología , Fosfatidilinositol 3-Quinasas/fisiología , Adenoviridae/fisiología , Animales , Transformación Celular Viral/genética , Células Cultivadas , Regulación Neoplásica de la Expresión Génica , Células HCT116 , Células HT29 , Células HeLa , Humanos , Proteínas Sustrato del Receptor de Insulina/genética , Proteína Oncogénica v-akt/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Unión Proteica , Ratas , Transducción de Señal
6.
Oncogene ; 26(53): 7467-79, 2007 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-17546052

RESUMEN

C-terminal-binding protein interacting protein (CtIP) was first isolated as a binding partner of C-terminal-binding protein (CtBP). It is considered to contribute to the transcriptional repression and cell cycle regulatory properties of the retinoblastoma (Rb) family of proteins and to have a role in the cellular response to DNA damage. Here, we have shown that CtIP is a novel target for the adenovirus oncoprotein early region 1A (AdE1A). AdE1A associates with CtIP in both Ad5E1-transformed cells and Ad5-infected cells and binds directly in glutathione-S-transferase pull-down assays. Two binding sites have been mapped on Ad5E1A - the N-terminal alpha-helical region (residues 1-30) and conserved region 3 (CR3) - the transcriptional activation domain. CtIP can bind AdE1A and CtBP independently, raising the possibility that ternary complexes exist in Ad-transformed and -infected cells. Significantly, reduction of CtIP expression with small interfering RNAs results in reduction of the ability of a Gal4 DNA-binding domain-CR3 construct to transactivate a Gal 4-responsive luciferase reporter and this effect is reversed by reduction of CtBP expression. Therefore, in this model, CtIP acts as a transcriptional co-activator of AdE1A when dissociated from CtBP, through the action of AdE1A. These data are consistent with observations that CtIP expression is induced by AdE1A during viral infection and that reduction of CtIP expression with RNA interference can retard virus replication. In addition, AdE1A causes disruption of the CtIP/Rb complex during viral infection by its interaction with CtIP, possibly contributing to transcriptional derepression.


Asunto(s)
Proteínas E1A de Adenovirus/metabolismo , Proteínas Portadoras/metabolismo , Proteínas Nucleares/metabolismo , Adenoviridae/fisiología , Proteínas E1A de Adenovirus/biosíntesis , Proteínas E1A de Adenovirus/genética , Infecciones por Adenovirus Humanos/genética , Infecciones por Adenovirus Humanos/metabolismo , Oxidorreductasas de Alcohol/metabolismo , Sitios de Unión , Neoplasias de la Mama/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Proteínas Portadoras/genética , Línea Celular Tumoral , Transformación Celular Viral , Proteínas de Unión al ADN/metabolismo , Endodesoxirribonucleasas , Humanos , Neoplasias Pulmonares/metabolismo , Proteínas Nucleares/genética , Unión Proteica , Estructura Terciaria de Proteína , Proteína de Retinoblastoma/metabolismo , Activación Transcripcional , Replicación Viral
7.
Br J Cancer ; 95(5): 555-60, 2006 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-16880778

RESUMEN

Adenovirus early region 1A (E1A) possesses potent transforming activity when expressed in concert with activated ras or E1B genes in in vitro tissue culture systems such as embryonic human retinal neuroepithelial cells or embryonic rodent epithelial and fibroblast cells. Early region 1A has thus been used extensively and very effectively as a tool to determine the molecular mechanisms that underlie the basis of cellular transformation. In this regard, roles for the E1A-binding proteins pRb, p107, p130, cyclic AMP response element-binding protein (CBP)/p300, p400, TRRAP and CtBP in cellular transformation have been established. However, the mechanisms by which E1A promotes transformation through interaction with these partner proteins are not fully delineated. In this review, we focus on recent advances in our understanding of CBP/p300 function, particularly with regard to its relationship to the anaphase-promoting complex/cyclosome E3 ubiquitin ligase, which has recently been shown to interact and affect the activity of CBP/p300 through interaction domains that are evolutionarily conserved in E1A.


Asunto(s)
Proteínas E1A de Adenovirus/fisiología , Proteína de Unión a CREB/metabolismo , Transformación Celular Neoplásica , Ubiquitina-Proteína Ligasas/metabolismo , Factores de Transcripción p300-CBP/metabolismo , Animales , Genes APC , Humanos , Roedores
9.
EMBO J ; 19(17): 4759-73, 2000 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-10970867

RESUMEN

We have identified the N-terminus of adenovirus early region 1A (AdE1A) as a region that can regulate the 26S proteasome. Specifically, in vitro and in vivo co-precipitation studies have revealed that the 19S regulatory components of the proteasome, Sug1 (S8) and S4, bind through amino acids (aa) 4-25 of Ad5 E1A. In vivo expression of wild-type (wt) AdE1A, in contrast to the N-terminal AdE1A mutant that does not bind the proteasome, reduces ATPase activity associated with anti-S4 immunoprecipitates relative to mock-infected cells. This reduction in ATPase activity correlates positively with the ability of wt AdE1A, but not the N-terminal deletion mutant, to significantly reduce the ability of HPV16 E6 to target p53 for ubiquitin-mediated proteasomal degradation. AdE1A/proteasomal complexes are present in both the cytoplasm and the nucleus, suggesting that AdE1A interferes with both nuclear and cytoplasmic proteasomal degradation. We have also demonstrated that wt AdE1A and the N-terminal AdE1A deletion mutant are substrates for proteasomal-mediated degradation. AdE1A degradation is not, however, mediated through ubiquitylation, but is regulated through phosphorylation of residues within a C-terminal PEST region (aa 224-238).


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteínas E1A de Adenovirus/metabolismo , Proteínas Portadoras/metabolismo , Péptido Hidrolasas/metabolismo , Complejo de la Endopetidasa Proteasomal , Proteínas Represoras , Factores de Transcripción , ATPasas Asociadas con Actividades Celulares Diversas , Adenosina Trifosfatasas/metabolismo , Sitios de Unión , ADN Helicasas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular , Proteínas con Dominio LIM , Proteínas Oncogénicas Virales/metabolismo , Fosforilación , Pruebas de Precipitina , Especificidad por Sustrato , Ubiquitinas/metabolismo
10.
Oncogene ; 19(3): 452-62, 2000 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-10656694

RESUMEN

The adenovirus early region 1B (Ad E1B) genes have no transforming capability of their own but markedly increase the transformation frequency of Ad E1A following co-transfection into mammalian cells. The larger E1B proteins of both Ad2/5 and Ad12 bind to p53 and inhibit its ability to transcriptionally activate other genes. We have previously demonstrated that synthetic peptides identical to the binding sites for p53 on both the Ad2 and Ad12 E1B proteins will disrupt the interaction in vivo and in vitro. In the work presented here we have examined the effects of complex dissociation on Ad E1-transformed human cells. It has been shown, using confocal microscopy, that when the peptide identical to the p53 binding site was added to Ad5 E1-transformed cells it initally located in the cytoplasmic dense bodies where it caused disruption of the p53/E1B complex. Peptide and p53 then translocated to the nucleus. In Ad12 E1-transformed cells the peptide localized in the nucleus directly and there caused a reorganization of p53 staining from a highly organized, 'flecked' distribution to one in which nuclear staining was homogeneous and diffuse. Peptides added to either Ad5 E1 or Ad12 E1 transformed cells resulted in the release of transcriptionally active p53. Interestingly, the level of p53 then fell presumably as a result of proteasomal action - this was probably a reflection of the short half-life of 'free' (i.e. dissociated) p53 compared to that of the bound protein. Free p53 did not cause apoptosis in target cells probably due to the presence of the smaller (19K) E1B proteins. However, addition of peptide leads to a significant reduction in cell growth rate. We have further demonstrated that a significant proportion of those cells which had taken up peptide had ceased DNA synthesis, probably due to a p53-induced cell cycle arrest. The role of the larger EIB protein during transformation is considered in view of these data.


Asunto(s)
Adenoviridae/fisiología , Proteínas E1B de Adenovirus/fisiología , Transformación Celular Neoplásica , Proteína p53 Supresora de Tumor/fisiología , Secuencia de Aminoácidos , Animales , Apoptosis , División Celular , Línea Celular , Humanos , Datos de Secuencia Molecular
11.
J Virol ; 73(3): 2074-83, 1999 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-9971789

RESUMEN

Recent reports suggest that an early region 1B (E1B) 55, 000-molecular-weight polypeptide (55K)-null adenovirus type 5 (Ad5) mutant (dl1520) can replicate to the same extent as wild-type (wt) Ad5 in cells either deficient or mutated in p53, implicating p53 in limiting viral replication in vivo. In contrast, we show here that the replicative capacity of Ad5 dl1520 is wholly independent of host cell p53 status, as is the replicative capacity of comparable Ad12 E1B 54K-null adenoviruses (Ad12 dl620 and Ad12 hr703). Furthermore, we show that there is no requirement for complex formation between p53 and Ad5 E1B 55K or Ad12 E1B 54K for a productive infection, such that wt Ad5 and wt Ad12 will both replicate in cells which are null for p53. In addition, we find that these Ad5 and Ad12 mutant viruses induce S phase irrespective of the p53 status of the cell and that, therefore, S-phase induction does not correlate with the replicative capacity of the virus. Interestingly, the replicative capacities of the large E1B-null adenoviruses correlated positively with the ability to express E1B 19K and were related to the ability to repress premature adenovirus-induced apoptosis. Infection of primary human cells indicated that Ad5 dl1520, wt Ad5, and wt Ad12 replicated better in cycling normal human skin fibroblasts (HSFs) than in quiescent HSFs. Thus, the cell cycle status of the host cell, upon infection, also influences viral yield.


Asunto(s)
Adenoviridae/fisiología , Proteínas E1B de Adenovirus/fisiología , Proteína p53 Supresora de Tumor/fisiología , Replicación Viral , Apoptosis , Fibroblastos/virología , Humanos , Fase S
12.
Oncogene ; 18(2): 449-58, 1999 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-9927201

RESUMEN

Adenovirus early region 1A (Ad E1A) is a multifunctional protein which is essential for adenovirus-mediated transformation and oncogenesis. Whilst E1A is generally considered to exert its influence on recipient cells through regulation of transcription it also increases the level of cellular p53 by increasing the protein half-life. With this in view, we have investigated the relationship of Ad E1A to the proteasome, which is normally responsible for degradation of p53. Here we have shown that both Ad5 and Ad12 E1A 12S and 13S proteins can be co-immunoprecipitated with proteasomes and that the larger Ad12 E1A protein binds strongly to at least three components of the 26S but not 20S proteasome. One of these interacting species has been identified as mammalian SUGI, a proteasome regulatory component which also plays a role in the cell as a mediator of transcription. In vitro assays have demonstrated a direct interaction between Ad12 E1A 13S protein and mouse SUGI. Following infection of human cells with Ad5 wt and Ad5 mutants with lesions in the E1A gene it has been shown that human SUG1 can be co-immunoprecipitated with full-length E1A and with E1A carrying a deletion in conserved region 1 which is the region considered to be responsible for increased expression of p53. We have concluded therefore that Ad EIA binds strongly to SUGI but that this interaction is not responsible for inhibition of proteasome activity. This is consistent with the observation that purified Ad12 E1A inhibits the activity of the purified 20S but not 26S proteasomes. We have also demonstrated that SUGI can be co-immunoprecipitated with SV40 T and therefore we suggest that this may represent a common interaction of transforming proteins of DNA tumour viruses.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteínas E1A de Adenovirus/metabolismo , Proteínas Portadoras/metabolismo , Cisteína Endopeptidasas/metabolismo , Complejos Multienzimáticos/metabolismo , Factores de Transcripción , ATPasas Asociadas con Actividades Celulares Diversas , Antígenos Transformadores de Poliomavirus/metabolismo , Línea Celular Transformada , Regulación de la Expresión Génica , Genes p53 , Humanos , Péptidos y Proteínas de Señalización Intracelular , Proteínas con Dominio LIM , Complejo de la Endopetidasa Proteasomal , Unión Proteica , Células Tumorales Cultivadas
13.
Virology ; 244(2): 330-42, 1998 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-9601504

RESUMEN

It has previously been shown that following viral infection, Ad5 E1A induces cell cycle progression of quiescent rodent cells, leading to DNA synthesis and mitosis. Here we have examined the effect of Ad12 E1A on the cell cycle characteristics of human cells. Human tumor (A549, KB, and HeLa) cells were infected with Ad12 d/620, a mutant virus which has a lesion in the E1B gene and essentially expresses only E1A. These infected cells progressed from being largely in G1 into S phase, where they arrested. Even up to 96 h postinfection (p.i.) the cells remained blocked in S phase. DNA synthesis did, however, proceed in Ad12 d/620-infected cells, giving rise to multiple copies of cellular DNA. Similar results were obtained when primary human skin fibroblasts were infected, although the polyploidy was less marked. The expression of cyclins A, B1, and E in the tumor cells increased appreciably in response to E1A. In contrast, there was a dramatic reduction in the levels of cyclin D1 and D3. Increases in cyclin D1 expression could be detected at very late times p.i. In those cell lines expressing low levels of cdc2 and cdk2 an appreciable increase in expression was seen soon after Ad12 E1A could be detected. The elevated levels of cyclins A, B1, and E were associated with increased protein kinase activity directed against histone H1. An increase in cyclin D1-associated kinase activity against Rb1 was also observed at late times. This deregulation of the cell cycle was not solely dependent on E1A inactivation of Rb, since similar effects were seen in Ad12 d/620-infected retinoblastoma (Y-79) cells, implicating p107 and p130 in E1A-mediated changes in cell cycle progression. We propose that the E1A-induced levels of cyclins A, B1, and E by Ad12 E1A in human cells may lead to an uncoupling of S phase from cell cycle progression.


Asunto(s)
Adenoviridae/patogenicidad , Proteínas E1A de Adenovirus/fisiología , Fase S , Adenoviridae/genética , Adenoviridae/fisiología , Proteínas E1A de Adenovirus/genética , Animales , Ciclo Celular , Línea Celular , Núcleo Celular/ultraestructura , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/metabolismo , ADN Viral/genética , ADN Viral/metabolismo , Células HeLa , Humanos , Mutación , Células Tumorales Cultivadas
14.
Virology ; 244(1): 230-42, 1998 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-9581794

RESUMEN

Adenovirus 12 early region 1A (Ad12 E1A) was expressed in Escherichia coli. Protein was purified in good yield in the presence of 8 M urea and then renatured by dialysis against dilute NH4HCO3 buffer. The affinity of this protein for pRb, C-terminal binding protein (CtBP), TATA binding protein (TBP), and SUG1 was similar to, or greater than, that of Ad12 E1A prepared by immunoaffinity chromatography under nondenaturing conditions. While the binding of the 266- and 235-amino-acid (aa) E1A components to TBP showed similar characteristics the larger E1A protein had a higher affinity for CtBP, pRb, and SUG1. Using nuclear magnetic resonance (NMR) spectroscopy it was shown that structural perturbations occurred in the 266-aa protein in the presence of Zn2+ consistent with binding--no such changes were seen for the 235-aa protein. Limited proteolysis of the 266- and 235-aa E1A proteins gave rise to comparable polypeptide products, suggesting overall similarities in structure. However, the different affinities of the 266- and 235-aa proteins for the partner proteins and the differences seen in the NMR spectra from the two proteins suggested structural differences.


Asunto(s)
Proteínas E1A de Adenovirus/metabolismo , Adenovirus Humanos/metabolismo , Proteínas E1A de Adenovirus/genética , Proteínas E1A de Adenovirus/aislamiento & purificación , Adenovirus Humanos/genética , Sitios de Unión , Endopeptidasas/metabolismo , Escherichia coli , Humanos , Unión Proteica , Desnaturalización Proteica , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/metabolismo , Zinc/metabolismo
16.
Biochem J ; 308 ( Pt 3): 965-73, 1995 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-8948457

RESUMEN

The mechanism by which thrombin and prothrombin control neurite retraction was studied in Ad12E1HER10 human neuroepithelial cells. Morphological changes in differentiated cells were apparent within minutes of the addition of very low concentrations of thrombin (3 pM). Higher concentrations (2 nM) of prothrombin were required to elicit a similar response. Doses of thrombin and prothrombin sufficient to cause neurite retraction stimulated protein tyrosine kinase activity. Protein tyrosine kinase activation also correlated positively with thrombin- and prothrombin-induced phosphoinositide 3-kinase activation and InsP6 dephosphorylation. However, thrombin-stimulated Ins(1,4,5)P3 generation and intracellular Ca2+ mobilization only occurred at concentrations in excess of those needed to induce retraction. No fluctuations in Ins(1,4,5)P3 were detected after stimulation with prothrombin, and no rapid synchronized release of Ca2+ was observed, even at very high concentrations. Prothrombin did, however, cause small oscillations in the intracellular Ca2+ concentration, similar to those produced by low concentrations of thrombin, after approximately 30 min. We conclude that prothrombin- and thrombin-induced neurite retractions are not dependent on PtdIns(4,5)P2 and Ca2+ mobilization, but are more probably mediated through an effector mechanism involving protein tyrosine kinase activation. No intracellular Ca2+ mobilization, protein tyrosine kinase activity or neurite retraction was observed after treatment of cells with proteolytically inactive mutant thrombin (S205-->A). Prothrombin-mediated intracellular Ca2+ mobilization and neurite retraction were inhibited by hirudin, which was shown to interact with thrombin but not prothrombin. It is concluded that cleavage of prothrombin to thrombin is a necessary prerequisite for biological activity on differentiated Ad12E1HER10 cells and that differences in agonist concentration are capable of coupling the thrombin receptor to different pathways within the cell.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Neuritas/efectos de los fármacos , Protrombina/farmacología , Trombina/farmacología , Western Blotting , Calcio/metabolismo , Células Cultivadas , Hirudinas/farmacología , Humanos , Fosfatos de Inositol/metabolismo , Fosfatidilinositol 3-Quinasas , Fosfatidilinositoles/metabolismo , Fosforilación , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Protrombina/metabolismo , Trombina/metabolismo
17.
Proc Biol Sci ; 247(1318): 27-33, 1992 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-1348118

RESUMEN

A homogeneous population of undifferentiated myeloid blast cells was purified from human fetal liver by rosette sedimentation of erythroblasts and macrophages, after coating these cells with monoclonal antibodies, followed by a cell elutriation step. The undifferentiated blast cells were maintained in culture, in a serum-free medium containing 1 mg l-1 inositol, by the presence of a high concentration of interleukin-3 (100 U ml-1). This allowed equilibrium labelling of cells with [2-3H]myo-inositol and analysis of the concentrations of inositol metabolites. The myeloid blast cells contained high concentrations of an unidentified inositol metabolite, possibly sn-glycero-3-phospho-1-inositol (GroPIns, 22 microM), inositol monophosphate (InsP, 16 microM), an unidentified inositol bisphosphate (InsP2, 9.4 microM), inositol pentakisphosphate (InsP5, 37 microM) and inositol hexakisphosphate (InsP6, 31 microM). These high concentrations are similar to those reported in the promyeloid cell line, HL60. Treatment of the blast cells with 10 nM phorbol myristate acetate (PMA) resulted in rapid differentiation of 48% of the cells towards monocytes. Notable changes in the levels of inositol metabolites included an increase in the putative GroPIns peak (to 73 microM) and decreases in the concentrations of InsP4 (from 4 microM to 1 microM) and InsP5 (to 21 microM). These changes in response to PMA, with the exception of the rise in the putative GroPIns, are similar to those reported in HL60 cells undergoing monocyte differentiation. These observations suggest that the abundant inositol polyphosphates may have an as yet unknown role in myeloid differentiation.


Asunto(s)
Diferenciación Celular , Fosfatos de Inositol/metabolismo , Inositol/metabolismo , Hígado/embriología , Monocitos/citología , Aborto Espontáneo , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Cromatografía Líquida de Alta Presión , Femenino , Feto , Humanos , Fosfatos de Inositol/aislamiento & purificación , Hígado/citología , Hígado/efectos de los fármacos , Monocitos/efectos de los fármacos , Embarazo , Acetato de Tetradecanoilforbol/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...