Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
Add more filters










Publication year range
1.
Oncogene ; 33(19): 2487-94, 2014 May 08.
Article in English | MEDLINE | ID: mdl-23792446

ABSTRACT

Whole chromosome instability (CIN) is a common feature of cancer cells and has been linked to increased tumor evolution and metastasis. Several studies have shown that the loss of the pRB tumor suppressor causes mitotic defects and chromosome mis-segregation. pRB is inactivated in many types of cancer and this raises the possibility that the loss of pRB may be a general cause of CIN in tumors. Paradoxically, retinoblastoma tumor cells have a relatively stable karyotype and currently the circumstances in which pRB inactivation causes CIN in human cancers are unclear. Here we utilize a fluorescence in situ hybridization-based approach to score numerical heterogeneity in chromosome copy number as a readout of CIN. Using this technique, we show that high levels of CIN correlate with the combined inactivation of pRB and p53 and that this association is evident in two independent panels of cancer cell lines. Retinoblastoma cell lines characteristically retain a wild-type TP53 gene, providing an opportunity to test the relevance of this functional relationship. We show that retinoblastoma cell lines display mitotic defects similar to those seen when pRB is depleted from non-transformed cells, but that the presence of wild-type p53 suppresses the accumulation of aneuploid cells. A similar synergy between pRB and p53 inactivation was observed in HCT116 cells. These results suggest that the loss of pRB promotes segregation errors, whereas loss of p53 allows tolerance and continued proliferation of the resulting, genomically unstable cancer cells. Hence, it is the cooperative effect of inactivation of both pRB and p53 tumor suppressor pathways that promotes CIN.


Subject(s)
Chromosomal Instability/genetics , Neoplasms/genetics , Retinoblastoma Protein/genetics , Retinoblastoma/genetics , Tumor Suppressor Protein p53/genetics , Animals , Cell Line, Tumor , Fluorescent Antibody Technique , Gene Knockdown Techniques , HCT116 Cells , Humans , In Situ Hybridization, Fluorescence , Neoplasms/metabolism , RNA, Small Interfering
2.
Proc Natl Acad Sci U S A ; 98(23): 13138-43, 2001 Nov 06.
Article in English | MEDLINE | ID: mdl-11687642

ABSTRACT

Cyclin E1 (formerly called cyclin E) and the recently described cyclin E2 belong to the family of E-type cyclins that operate during the G(1)/S phase progression in mammalian cells. The two E-cyclins share a catalytic partner, cyclin-dependent kinase 2 (CDK2), and activate their associated kinase activities at similar times during cell cycle progression. Despite these similarities, it is unknown whether the two proteins perform distinct functions, or, alternatively, they control S-phase entry of different cell types in a tissue-specific fashion. To start addressing in vivo functions of E-cyclins, we determined the expression pattern of cyclins E1 and E2 during normal mouse development. We found that the two E-cyclins showed very similar patterns of expression; both were expressed within the proliferating compartment during embryo development. Analyses of cells and tissues lacking members of the retinoblastoma (pRB) family of proteins revealed that the expression of both cyclins is controlled in a pRB-dependent, but p107- and p130-independent fashion, likely through the pRB-dependent E2F transcription factors. We also found that cyclins E1 and E2 are expressed at high levels in mouse breast tumors driven by the Myc oncogene. Last, we found that cyclin E2 is overexpressed in approximately 24% of analyzed human mammary carcinomas. Collectively these findings suggest that the expression of cyclins E1 and E2 is governed by similar molecular circuitry.


Subject(s)
Breast Neoplasms/genetics , Cyclin E/genetics , Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Animals , Blotting, Northern , Embryo, Mammalian/metabolism , Embryonic and Fetal Development/genetics , Gene Expression Regulation, Developmental/physiology , Gene Expression Regulation, Neoplastic/physiology , Humans , Mice , Retinoblastoma Protein/physiology , Stem Cells/metabolism
3.
Genes Dev ; 15(16): 2146-60, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11511545

ABSTRACT

E2F is a heterogenous transcription factor and its role in cell cycle control results from the integrated activities of many different E2F family members. Unlike mammalian cells, that have a large number of E2F-related genes, the Drosophila genome encodes just two E2F genes, de2f1 and de2f2. Here we show that de2f1 and de2f2 provide different elements of E2F regulation and that they have opposing functions during Drosophila development. dE2F1 and dE2F2 both heterodimerize with dDP and bind to the promoters of E2F-regulated genes in vivo. dE2F1 is a potent activator of transcription, and the loss of de2f1 results in the reduced expression of E2F-regulated genes. In contrast, dE2F2 represses the transcription of E2F reporters and the loss of de2f2 function results in increased and expanded patterns of gene expression. The loss of de2f1 function has previously been reported to compromise cell proliferation. de2f1 mutant embryos have reduced expression of E2F-regulated genes, low levels of DNA synthesis, and hatch to give slow-growing larvae. We find that these defects are due in large part to the unchecked activity of dE2F2, since they can be suppressed by mutation of de2f2. Examination of eye discs from de2f1; de2f2 double-mutant animals reveals that relatively normal patterns of DNA synthesis can occur in the absence of both E2F proteins. This study shows how repressor and activator E2Fs are used to pattern transcription and how the net effect of E2F on cell proliferation results from the interplay between two types of E2F complexes that have antagonistic functions.


Subject(s)
Cell Cycle Proteins , DNA-Binding Proteins , Drosophila Proteins , Transcription Factors/antagonists & inhibitors , Alleles , Animals , Animals, Genetically Modified , Cell Cycle , Drosophila/genetics , Drosophila/physiology , E2F Transcription Factors , E2F2 Transcription Factor , Eye , Gene Deletion , Gene Expression Regulation , Phenotype , Retinoblastoma Protein , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factors/physiology , Transcription, Genetic/physiology
4.
Clin Cancer Res ; 7(8): 2387-95, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11489817

ABSTRACT

PURPOSE: Analysis of tumor-derived genetic lesions has provided insights into molecular pathogenesis of human gliomas. Because these changes represent only one of several mechanisms that alter gene expression during tumorigenesis, it is likely that further information will be obtained from a careful analysis of important regulatory proteins present in these tumors. EXPERIMENTAL DESIGN: We have quantified the levels of key cell cycle/signaling proteins in 94 prospectively collected, meticulously preserved, "snap frozen" glioma specimens and have compared these levels with histopathological data and patient outcome. RESULTS: The results of these experiments confirm that the levels of wild-type tumor suppressor proteins, such as p53, pRB, PTEN, p14(ARF), and p16(INK4), are lost or severely reduced in most gliomas, and that epidermal growth factor receptor, 2human telomerase reverse transcriptase, and cyclin-dependent kinase 4 are overexpressed frequently and with a few exceptions, almost exclusively, in glioblastomas. In addition, we report frequent underexpression of E2F-1 (in 55% of gliomas) and cyclin E overexpression (in 26% of gliomas), which have not yet been reported on the genomic level. Several of these markers significantly correlated with histopathological grade, and the levels of five proteins showed significant association with patient outcome. In particular, overexpression of epidermal growth factor receptor, human telomerase reverse transcriptase, cyclin-dependent kinase 4, and cyclin E was largely restricted to glioblastomas and was significantly associated with reduced patient survivals. CONCLUSIONS: We conclude that the quantitation of cell cycle/signaling proteins from meticulously preserved glioma specimens provides further insights into the molecular pathogenesis of human gliomas and yields valuable prognostic information.


Subject(s)
Cell Cycle Proteins/analysis , Glioma/pathology , Proto-Oncogene Proteins , Tumor Suppressor Proteins , Blotting, Western , Cell Cycle Proteins/biosynthesis , Cyclin D1/analysis , Cyclin E/analysis , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase Inhibitor p16/analysis , Cyclin-Dependent Kinases/analysis , DNA-Binding Proteins , ErbB Receptors/analysis , Glioma/metabolism , Humans , PTEN Phosphohydrolase , Phosphoric Monoester Hydrolases/analysis , Prognosis , Proteins/analysis , Retinoblastoma Protein/analysis , Telomerase/analysis , Tumor Suppressor Protein p14ARF , Tumor Suppressor Protein p53/analysis
5.
J Neurosci ; 21(14): 5017-26, 2001 Jul 15.
Article in English | MEDLINE | ID: mdl-11438577

ABSTRACT

DNA damage has been implicated as one important initiator of cell death in neuropathological conditions such as stroke. Accordingly, it is important to understand the signaling processes that control neuronal death induced by this stimulus. Previous evidence has shown that the death of embryonic cortical neurons treated with the DNA-damaging agent camptothecin is dependent on the tumor suppressor p53 and cyclin-dependent kinase (CDK) activity and that the inhibition of either pathway alone leads to enhanced and prolonged survival. We presently show that p53 and CDKs are activated independently on parallel pathways. An increase in p53 protein levels, nuclear localization, and DNA binding that result from DNA damage are not affected by the inhibition of CDK activity. Conversely, no decrease in retinoblastoma protein (pRb) phosphorylation was observed in p53-deficient neurons that were treated with camptothecin. However, either p53 deficiency or the inhibition of CDK activity alone inhibited Bax translocation, cytochrome c release, and caspase-3-like activation. Taken together, our results indicate that p53 and CDK are activated independently and then act in concert to control Bax-mediated apoptosis.


Subject(s)
Cyclin-Dependent Kinases/metabolism , DNA Damage/physiology , Neurons/metabolism , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/physiology , Animals , Apoptosis/physiology , Camptothecin/pharmacology , Caspase 3 , Caspases/metabolism , Cell Survival , Cells, Cultured , Cyclin-Dependent Kinases/antagonists & inhibitors , Cytochrome c Group/metabolism , Enzyme Inhibitors/pharmacology , Mice , Mice, Knockout , Neurons/cytology , Neurons/drug effects , Phosphorylation/drug effects , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Retinoblastoma Protein/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Tumor Suppressor Protein p53/deficiency , bcl-2-Associated X Protein
6.
Proc Natl Acad Sci U S A ; 98(15): 8720-5, 2001 Jul 17.
Article in English | MEDLINE | ID: mdl-11447271

ABSTRACT

We have developed a yeast model system to address transcriptional repression by the retinoblastoma protein (pRB). When fused to the DNA-binding domain of Gal4p (DB-pRB), pRB can repress transcription of reporter genes containing Gal4p binding sites; the histone deacetylase activity encoded by yeast RPD3 is required for DB-pRB repression. Mutation of the LXCXE binding cleft in pRB, a region reported to be required for histone deacetylase recruitment, does not interfere with pRB-mediated repression. From these findings based on yeast experiments, we surmise that the small pocket region of pRB must contain an additional domain that confers histone deacetylase-dependent transcriptional repression. This hypothesis was verified by experiments examining pRB-dependent histone deacetylase association in mammalian cells. In addition to RPD3, repression by pRB in yeast requires MSI1, an ortholog of RbAp48, but not SIN3 or SAP30. By comparing the genetic requirements of DB-pRB repression in yeast to those of other DB-repressor fusions, we can suggest a mechanism by which pRB recruits histone deacetylase activity.


Subject(s)
DNA-Binding Proteins , Gene Expression Regulation , Histone Deacetylases/metabolism , Repressor Proteins/metabolism , Retinoblastoma Protein/metabolism , Saccharomyces cerevisiae Proteins , Binding Sites , Chromatin Assembly Factor-1 , Fungal Proteins/genetics , Fungal Proteins/metabolism , Histone Deacetylase 1 , Histone Deacetylases/genetics , Hydro-Lyases/genetics , Repressor Proteins/genetics , Retinoblastoma Protein/genetics , Saccharomyces cerevisiae , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic
7.
Adv Cancer Res ; 82: 1-54, 2001.
Article in English | MEDLINE | ID: mdl-11447760

ABSTRACT

Studies of the retinoblastoma gene (Rb) have shown that its protein product (pRb) acts to restrict cell proliferation, inhibit apoptosis, and promote cell differentiation. The frequent mutation of the Rb gene, and the functional inactivation of pRb in tumor cells, have spurred interest in the mechanism of pRb action. Recently, much attention has focused on pRb's role in the regulation of the E2F transcription factor. However, biochemical studies have suggested that E2F is only one of many pRb-targets and, to date, at least 110 cellular proteins have been reported to associate with pRb. The plethora of pRb-binding proteins raises several important questions. How many functions does pRb possess, which of these functions are important for development, and which contribute to tumor suppression? The goal of this review is to summarize the current literature of pRb-associated proteins.


Subject(s)
Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Genes, Retinoblastoma/physiology , Retinoblastoma Protein/metabolism , Transcription Factors/metabolism , Humans
8.
Mol Cell ; 7(4): 715-27, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11336696

ABSTRACT

Retinoblastoma (Rb) protein promotes cell survival after DNA damage. We show here that the LxCxE binding site in Rb mediates both cell survival and cell-cycle arrest after DNA damage. Replication factor C (RF-C) complex plays an important role in DNA replication. We describe a novel function of the large subunit of RF-C in promoting cell survival after DNA damage. RF-Cp145 contains an LxCxE motif, and mutation of this motif abolishes the protective effect of RF-Cp145. The inability of wild-type RF-Cp145 to promote cell survival in Rb-null cells is rescued by Rb but not by Rb mutants defective in binding LxCxE proteins. RF-C thus enhances cell survival after DNA damage in an Rb-dependent manner.


Subject(s)
DNA Damage/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Homeodomain Proteins , Proto-Oncogene Proteins c-bcl-2 , Repressor Proteins , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Saccharomyces cerevisiae Proteins , Amino Acid Motifs , Animals , Binding Sites/physiology , COS Cells , Cell Cycle/genetics , Cell Survival/genetics , Cell Survival/radiation effects , DNA Helicases , DNA-Binding Proteins/chemistry , Female , Histone Deacetylase 1 , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Minor Histocompatibility Antigens , Mutagenesis/physiology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Replication Protein C , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection , Tumor Cells, Cultured , Ultraviolet Rays , Uterine Cervical Neoplasms
9.
Exp Cell Res ; 264(1): 135-47, 2001 Mar 10.
Article in English | MEDLINE | ID: mdl-11237530

ABSTRACT

p107 and p130 were originally identified as targets of the transforming domains of viral oncoproteins encoded by small DNA tumor viruses. Together with pRB, the protein product of the retinoblastoma gene (Rb), p107 and p130 represent a family of closely related proteins that play critical roles in the regulation of cell proliferation. p107, p130, and pRB are transcriptional regulators whose activities are coupled to the cell cycle. Each of these proteins associates with E2F and is directly regulated by phosphorylation by cyclin-dependent kinases. In vivo studies of p107 and p130 function have revealed that their roles overlap extensively with one another and with pRB. In addition, the analysis of mice (and cell lines derived from these animals) deficient in these proteins shows that the individual members of this family harbor distinct functions that, at present, are poorly understood. The characterization of tumor cells continues to emphasize the important and somewhat unique role of pRB in tumor suppression, and the evidence linking the specific inactivation of p107 or p130 to tumor development remains quite limited. In this review we summarize the biochemical and functional properties of p107 and p130, and we compare and contrast these properties to those of pRB.


Subject(s)
Neoplasms/genetics , Nuclear Proteins/physiology , Phosphoproteins/physiology , Proteins , Animals , Cell Cycle , Cell Differentiation , Humans , Mice , Nuclear Proteins/chemistry , Phosphoproteins/chemistry , Retinoblastoma Protein/chemistry , Retinoblastoma Protein/physiology , Retinoblastoma-Like Protein p107 , Retinoblastoma-Like Protein p130
10.
Genes Dev ; 14(22): 2855-68, 2000 Nov 15.
Article in English | MEDLINE | ID: mdl-11090133

ABSTRACT

Using methods that conserve nuclear architecture, we have reanalyzed the spatial organization of the initiation of mammalian DNA synthesis. Contrary to the commonly held view that replication begins at hundreds of dispersed nuclear sites, primary fibroblasts initiate synthesis in a limited number of foci that contain replication proteins, surround the nucleolus, and overlap with previously identified internal lamin A/C structures. These foci are established in early G(1)-phase and also contain members of the retinoblastoma protein family. Later, in S-phase, DNA replication sites distribute to regions located throughout the nucleus. As this progression occurs, association with the lamin structure and pRB family members is lost. A similar temporal progression is found in all the primary cells we have examined but not in most established cell lines, indicating that the immortalization process modifies spatial control of DNA replication. These findings indicate that in normal mammalian cells, the onset of DNA synthesis is coordinately regulated at a small number of previously unrecognized perinucleolar sites that are selected in early G(1)-phase.


Subject(s)
Cell Nucleus/ultrastructure , DNA Replication , 3T3 Cells , Animals , Cell Nucleolus/ultrastructure , Cells, Cultured , Culture Techniques/methods , G1 Phase , Humans , Lamin Type A , Lamins , Mice , Nuclear Proteins/isolation & purification , Retinoblastoma Protein/isolation & purification , S Phase
11.
EMBO J ; 19(20): 5376-86, 2000 Oct 16.
Article in English | MEDLINE | ID: mdl-11032805

ABSTRACT

Mutations in ebi were isolated as enhancers of an over-proliferation phenotype generated by elevated E2F/DP activity in the Drosophila eye. ebi alleles also strongly suppress a phenotype caused by the cyclin-dependent kinase inhibitor p21, restoring S phases in the second mitotic wave of the developing eye disk. ebi mutant embryos display ectopic S phases within the peripheral nervous system and central nervous system at a time in development when neuronal precursor cells would normally begin to differentiate. Consistent with this, we find that ebi mutants have a reduced capacity to undergo neuronal differentiation, that Ebi physically interacts with Sina and phyllopod, and that Ebi promotes Ttk88 degradation in vitro and in S2 cells. Ectopic expression of Ttk88 inhibited differentiation in embryos and eye discs; however, this block to differentiation was insufficient to promote S phase entry in either of the situations where ebi mutations gave this effect. We conclude that Ebi has two distinct functions; it promotes the degradation of a repressor of neuronal differentiation (Ttk88), and has a second independent function that limits S phase entry.


Subject(s)
Cell Cycle Proteins , Cell Cycle , Drosophila Proteins , Drosophila melanogaster/cytology , Drosophila melanogaster/embryology , Eye/embryology , GTP-Binding Proteins , Insect Proteins/metabolism , Neurons/metabolism , Alleles , Amino Acid Sequence , Animals , Cell Differentiation , Drosophila melanogaster/genetics , E2F2 Transcription Factor , Eye/ultrastructure , Gene Expression Regulation, Developmental , Genes, Dominant/genetics , Insect Proteins/chemistry , Insect Proteins/genetics , Microscopy, Electron, Scanning , Molecular Sequence Data , Nervous System/cytology , Nervous System/embryology , Phenotype , Protein Binding , Protein Structure, Tertiary , Repressor Proteins/chemistry , Repressor Proteins/genetics , Repressor Proteins/metabolism , S Phase , Sequence Alignment , Suppression, Genetic/genetics , Transcription Factors/metabolism , Ubiquitins/metabolism
12.
Mol Cell ; 6(3): 737-42, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11030353

ABSTRACT

Analysis of tumor-derived mutations has led to the suggestion that p16INK4a, cyclin D1, cdk4, and the retinoblastoma protein (pRB) are components of a regulatory pathway that is inactivated in most tumor cells. Cell cycle arrest induced by p16INK4a, an inhibitor of cyclin D-dependent kinases, requires pRB, and it has been proposed that this G1 arrest is mediated by pRB-E2F repressor complexes. By comparing the properties of primary mouse embryonic fibroblasts specifically lacking pRB-family members, we find that pRB is insufficient for a p16INK4a-induced arrest. In addition to pRB, a second function provided by either p107 or p130, two pRB-related proteins, is required for p16INK4a to block DNA synthesis. We infer that p16INK4a-induced arrest is not mediated exclusively by pRB, but depends on the nonredundant functions of at least two pRB-family members.


Subject(s)
Carrier Proteins/genetics , G1 Phase/physiology , Gene Expression Regulation, Neoplastic/physiology , Proteins , Proto-Oncogene Proteins , Retinoblastoma Protein/genetics , Animals , Blotting, Western , Carrier Proteins/analysis , Cells, Cultured , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase Inhibitor p16 , Cyclin-Dependent Kinases/analysis , Fetus/cytology , Fibroblasts/chemistry , Fibroblasts/cytology , Mice , Mice, Knockout , Nuclear Proteins/genetics , Phosphoproteins/genetics , Retinoblastoma-Like Protein p107 , Retinoblastoma-Like Protein p130 , S Phase/physiology
13.
J Biol Chem ; 275(16): 11553-60, 2000 Apr 21.
Article in English | MEDLINE | ID: mdl-10766769

ABSTRACT

Although B-amyloid (AB) is suggested to play an important role in Alzheimer's disease, the mechanisms that control AB-evoked toxicity are unclear. We demonstrated previously that the cell cycle-related cyclin-dependent kinase 4/6/retinoblastoma protein pathway is required for AB-mediated death. However, the downstream target(s) of this pathway are unknown. We show here that neurons lacking E2F1, a transcription factor regulated by the retinoblastoma protein, are significantly protected from death evoked by AB. Moreover, p53 deficiency does not protect neurons from death, indicating that E2F1-mediated death occurs independently of p53. Neurons protected by E2F1 deficiency have reduced Bax-dependent caspase 3-like activity. However, protection afforded by E2F1, Bax, or caspase 3 deficiency is transient. In the case of E2F1, but not with Bax or caspase 3 deficiency, delayed death is accompanied by DEVD-AFC cleavage activity. Taken together, these results demonstrate the required role of E2F1, Bax, and caspase 3 in AB evoked death, but also suggest the participation of elements independent of these apoptosis regulators.


Subject(s)
Amyloid beta-Peptides/pharmacology , Apoptosis , Carrier Proteins , Caspases/metabolism , Cell Cycle Proteins/physiology , Cerebral Cortex/cytology , DNA-Binding Proteins/physiology , Neurons/physiology , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins/metabolism , Transcription Factors/physiology , Animals , Caspase 3 , Cells, Cultured , Cerebral Cortex/drug effects , Coumarins/pharmacology , E2F Transcription Factors , E2F1 Transcription Factor , Enzyme Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/drug effects , Oligopeptides/pharmacology , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1 , Tumor Suppressor Protein p53/physiology , bcl-2-Associated X Protein
14.
Mol Cell Biol ; 20(10): 3715-27, 2000 May.
Article in English | MEDLINE | ID: mdl-10779361

ABSTRACT

The pocket domain of pRB is required for pRB to arrest the cell cycle. This domain was originally defined as the region of the protein that is necessary and sufficient for pRB's interaction with adenovirus E1A and simian virus s40 large T antigen. These oncoproteins, and other pRB-binding proteins that are encoded by a variety of plant and animal viruses, use a conserved LXCXE motif to interact with pRB. Similar sequences have been identified in multiple cellular pRB-binding proteins, suggesting that the viruses have evolved to target a highly conserved binding site of pRB that is critical for its function. Here we have constructed a panel of pRB mutants in which conserved amino acids that are predicted to make close contacts with an LXCXE peptide were altered. Despite the conservation of the LXCXE binding site throughout evolution, pRB mutants that lack this site are able to induce a cell cycle arrest in a pRB-deficient tumor cell line. This G(1) arrest is overcome by cyclin D-cdk4 complexes but is resistant to inactivation by E7. Consequently, mutants lacking the LXCXE binding site were able to induce a G(1) arrest in HeLa cells despite the expression of HPV-18 E7. pRB mutants lacking the LXCXE binding site are defective in binding to adenovirus E1A and human papillomavirus type 16 E7 protein but exhibit wild-type binding to E2F or DP, and they retain the ability to interact with CtIP and HDAC1, two transcriptional corepressors that contain LXCXE-like sequences. Consistent with these observations, the pRB mutants are able to actively repress transcription. These observations suggest that viral oncoproteins depend on the LXCXE-binding site of pRB for interaction to a far greater extent than cellular proteins that are critical for cell cycle arrest or transcriptional repression. Mutation of this binding site allows pRB to function as a cell cycle regulator while being resistant to inactivation by viral oncoproteins.


Subject(s)
Carrier Proteins , Cell Cycle Proteins/metabolism , DNA-Binding Proteins , Oncogene Proteins, Viral/metabolism , Proto-Oncogene Proteins , Retinoblastoma Protein/metabolism , Adenovirus E1A Proteins , Amino Acid Sequence , Binding Sites , Cell Cycle , Conserved Sequence , Cyclin D , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinases , Cyclins/metabolism , E2F Transcription Factors , Gene Expression Regulation , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Papillomavirus E7 Proteins , Protein Binding , Retinoblastoma Protein/genetics , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1 , Transcription Factors/metabolism , Tumor Cells, Cultured
15.
J Chromatogr A ; 868(2): 305-12, 2000 Feb 04.
Article in English | MEDLINE | ID: mdl-10701680

ABSTRACT

The paper published by Ghaoui and Rothman [J. High Resolut. Chromatogr. 15 (1992) 36] and particularly its Fig. 5, is further considered here because it contains the germ of an idea of how to measure improvements to a chromatographic method, and how to define the goal of perfection in terms of "zero defects" as required by quality assurance schemes. From this, a new role emerges for signal averaging in capillary chromatography: a role to quantify and measure method improvements, and one which can be generally applied to measure improvements in instrument design too.


Subject(s)
Chromatography/standards , Sensitivity and Specificity
16.
Genetics ; 153(1): 275-87, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10471712

ABSTRACT

The activity of the E2F transcription factor is regulated in part by pRB, the protein product of the retinoblastoma tumor suppressor gene. Studies of tumor cells show that the p16(ink4a)/cdk4/cyclin D/pRB pathway is mutated in most forms of cancer, suggesting that the deregulation of E2F, and hence the cell cycle, is a common event in tumorigenesis. Extragenic mutations that enhance or suppress E2F activity are likely to alter cell-cycle control and may play a role in tumorigenesis. We used an E2F overexpression phenotype in the Drosophila eye to screen for modifiers of E2F activity. Coexpression of dE2F and its heterodimeric partner dDP in the fly eye induces S phases and cell death. We isolated 33 enhancer mutations of this phenotype by EMS and X-ray mutagenesis and by screening a deficiency library collection. The majority of these mutations sorted into six complementation groups, five of which have been identified as alleles of brahma (brm), moira (mor) osa, pointed (pnt), and polycephalon (poc). osa, brm, and mor encode proteins with homology to SWI1, SWI2, and SWI3, respectively, suggesting that the activity of a SWI/SNF chromatin-remodeling complex has an important impact on E2F-dependent phenotypes. Mutations in poc also suppress phenotypes caused by p21(CIP1) expression, indicating an important role for polycephalon in cell-cycle control.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , Cell Cycle , DNA-Binding Proteins , Drosophila Proteins , Drosophila melanogaster/genetics , Genes, Insect , Trans-Activators , Transcription Factors/metabolism , Animals , Cell Death , Dimerization , Drosophila melanogaster/anatomy & histology , Drosophila melanogaster/embryology , Drosophila melanogaster/growth & development , E2F Transcription Factors , Eye/growth & development , Eye/metabolism , Eye/ultrastructure , Genes, Dominant/genetics , Genes, cdc/genetics , Genes, cdc/physiology , Genetic Complementation Test , Insect Proteins/genetics , Insect Proteins/physiology , Microscopy, Electron, Scanning , Mutation , Phenotype , Retinoblastoma Protein , Retinoblastoma-Binding Protein 1 , Sequence Homology, Amino Acid , Suppression, Genetic/genetics , Transcription Factors/genetics
17.
Mol Cell Biol ; 19(9): 6458-68, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10454591

ABSTRACT

Complexes between the retinoblastoma protein (pRb) and the transcription factor E2F-1 are thought to be important for regulating cell proliferation. We have shown previously that the E7 oncoprotein from human papillomavirus type 16, dependent upon its binding to pRb proteins, induces proliferation, disrupts differentiation, and induces apoptosis when expressed in the differentiating, or fiber, cells of the ocular lenses in transgenic mice. Mice that carry a null mutation in E2F-1 do not exhibit any defects in proliferation and differentiation in the lens. By examining the lens phenotype in mice that express E7 on an E2F-1 null background, we now show genetic evidence that E7's ability to alter the fate of fiber cells is partially dependent on E2F-1. On the other hand, E2F-1 status does not affect E7-induced proliferation in the undifferentiated lens epithelium. These data provide genetic evidence that E2F-1, while dispensible for normal fiber cell differentiation, is one mediator of E7's activity in vivo and that the requirement for E2F-1 is context dependent. These data suggest that an important role for pRb-E2F-1 complex during fiber cell differentiation is to negatively regulate cell cycle progression, thereby allowing completion of the differentiation program to occur.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , DNA-Binding Proteins , Lens, Crystalline/cytology , Nuclear Proteins/physiology , Oncogene Proteins, Viral/genetics , Transcription Factors/physiology , Animals , Animals, Newborn , Apoptosis , Cell Cycle , Cell Differentiation , Cell Division , DNA/biosynthesis , E2F Transcription Factors , E2F1 Transcription Factor , Female , Humans , Lens, Crystalline/embryology , Lens, Crystalline/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Nuclear Proteins/genetics , Oncogene Proteins, Viral/metabolism , Papillomaviridae/genetics , Papillomavirus E7 Proteins , Pregnancy , Protein Binding , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1 , Transcription Factors/genetics
18.
Mol Carcinog ; 25(4): 295-303, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10449036

ABSTRACT

The E2F family of transcription factors regulates the expression of genes needed for DNA synthesis and cell-cycle control. However, the individual contributions of the different E2F family members in regulating proliferation in various tissues have not been well characterized. Mouse liver is an excellent system for investigating proliferation because its growth state can be experimentally manipulated. As observed in cell culture systems, E2F1 protein is present at low levels in the quiescent liver, with an increase in expression during proliferation. Therefore, we expected that E2F1 may play an important role in cell-growth control during periods of robust proliferation. Using E2F1-nullizygous mice, we performed partial hepatectomies to investigate the role of E2F1 in the synchronous proliferation of adult hepatocytes. We found that E2F1 deficiency resulted in only minor changes in gene expression and that the timing of liver regeneration was not altered in E2F1 nullizygous mice. E2F1 has displayed properties of both a tumor suppressor and an oncogene in different model systems. Therefore, we investigated the role of E2F1 in rapidly growing liver tumor cells in strains of mice that have high (C3H/HeJ) and low (C57BL/6J) rates of hepatocarcinogenesis. We observed no significant differences in the number of liver tumors that developed after diethylnitrosamine treatment of wild type versus E2F1-nullizygous mice. We suggest that abundant levels of E2F4 in the mouse liver compensate for loss of E2F1.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , DNA-Binding Proteins , Liver Neoplasms, Experimental/genetics , Liver Regeneration/genetics , Transcription Factors/genetics , Animals , CDC2 Protein Kinase/genetics , Cell Division/genetics , Cell Transformation, Neoplastic/genetics , E2F Transcription Factors , E2F1 Transcription Factor , E2F4 Transcription Factor , Hepatectomy , Liver Neoplasms, Experimental/pathology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , RNA, Messenger/genetics , Retinoblastoma-Binding Protein 1 , Tetrahydrofolate Dehydrogenase/genetics , Transcription Factor DP1
20.
EMBO J ; 18(4): 916-25, 1999 Feb 15.
Article in English | MEDLINE | ID: mdl-10022834

ABSTRACT

The first appearance of G1 during Drosophila embryogenesis, at cell cycle 17, is accompanied by the down-regulation of E2F-dependent transcription. Mutant alleles of rbf were generated and analyzed to determine the role of RBF in this process. Embryos lacking both maternal and zygotic RBF products show constitutive expression of PCNA and RNR2, two E2F-regulated genes, indicating that RBF is required for their transcriptional repression. Despite the ubiquitous expression of E2F target genes, most epidermal cells enter G1 normally. Rather than pausing in G1 until the appropriate time for cell cycle progression, many of these cells enter an ectopic S-phase. These results indicate that the repression of E2F target genes by RBF is necessary for the maintenance but not the initiation of a G1 phase. The phenotype of RBF-deficient embryos suggests that rbf has a function that is complementary to the roles of dacapo and fizzy-related in the introduction of G1 during Drosophila embryogenesis.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , DNA-Binding Proteins , Drosophila Proteins , Drosophila/embryology , Gene Expression Regulation, Developmental/genetics , Trans-Activators , Transcription Factors/genetics , Animals , Cell Cycle/genetics , Cyclin E , Cyclin-Dependent Kinases/metabolism , E2F Transcription Factors , Embryonic Development , Eye/embryology , Eye/ultrastructure , G1 Phase/genetics , Gene Deletion , In Situ Hybridization , Microscopy, Electron, Scanning , Mutation/genetics , Phenotype , Repressor Proteins/genetics , Retinoblastoma Protein , Retinoblastoma-Binding Protein 1 , Transcription Factors/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...