Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
2.
Leukemia ; 30(6): 1388-98, 2016 06.
Article in English | MEDLINE | ID: mdl-26876596

ABSTRACT

TEN-ELEVEN-TRANSLOCATION-2 (TET2) and DNA-METHYLTRANSFERASE-3A (DNMT3A), both encoding proteins involved in regulating DNA methylation, are mutated in hematological malignancies affecting both myeloid and lymphoid lineages. We previously reported an association of TET2 and DNMT3A mutations in progenitors of patients with angioimmunoblastic T-cell lymphomas (AITL). Here, we report on the cooperative effect of Tet2 inactivation and DNMT3A mutation affecting arginine 882 (DNMT3A(R882H)) using a murine bone marrow transplantation assay. Five out of eighteen primary recipients developed hematological malignancies with one mouse developing an AITL-like disease, two mice presenting acute myeloid leukemia (AML)-like and two others T-cell acute lymphoblastic leukemia (T-ALL)-like diseases within 6 months following transplantation. Serial transplantations of DNMT3A(R882H) Tet2(-/-) progenitors led to a differentiation bias toward the T-cell compartment, eventually leading to AITL-like disease in 9/12 serially transplanted recipients. Expression profiling suggested that DNMT3A(R882H) Tet2(-/-) T-ALLs resemble those of NOTCH1 mutant. Methylation analysis of DNMT3A(R882H) Tet2(-/-) T-ALLs showed a global increase in DNA methylation affecting tumor suppressor genes and local hypomethylation affecting genes involved in the Notch pathway. Our data confirm the transformation potential of DNMT3A(R882H) Tet2(-/-) progenitors and represent the first cooperative model in mice involving Tet2 inactivation driving lymphoid malignancies.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation/genetics , DNA-Binding Proteins/genetics , Lymphoproliferative Disorders/genetics , Mutation , Proto-Oncogene Proteins/genetics , Animals , Cell Differentiation , DNA Methyltransferase 3A , Dioxygenases , Genes, Tumor Suppressor , Lymphoproliferative Disorders/etiology , Mice , Receptors, Notch/genetics
3.
Oncogene ; 35(33): 4368-78, 2016 08 18.
Article in English | MEDLINE | ID: mdl-26804170

ABSTRACT

Bruton's tyrosine kinase (BTK) is essential for B-cell proliferation/differentiation and it is generally believed that its expression and function are limited to bone marrow-derived cells. Here, we report the identification and characterization of p65BTK, a novel isoform abundantly expressed in colon carcinoma cell lines and tumour tissue samples. p65BTK protein is expressed, through heterogeneous nuclear ribonucleoprotein K (hnRNPK)-dependent and internal ribosome entry site-driven translation, from a transcript containing an alternative first exon in the 5'-untranslated region, and is post-transcriptionally regulated, via hnRNPK, by the mitogen-activated protein kinase (MAPK) pathway. p65BTK is endowed with strong transforming activity that depends on active signal-regulated protein kinases-1/2 (ERK1/2) and its inhibition abolishes RAS transforming activity. Accordingly, p65BTK overexpression in colon cancer tissues correlates with ERK1/2 activation. Moreover, p65BTK inhibition affects growth and survival of colon cancer cells. Our data reveal that BTK, via p65BTK expression, is a novel and powerful oncogene acting downstream of the RAS/MAPK pathway and suggest that its targeting may be a promising therapeutic approach.


Subject(s)
Cell Transformation, Neoplastic , Colonic Neoplasms/pathology , Protein-Tyrosine Kinases/physiology , ras Proteins/physiology , 5' Untranslated Regions/physiology , Agammaglobulinaemia Tyrosine Kinase , Cell Line, Tumor , Colonic Neoplasms/enzymology , Heterogeneous-Nuclear Ribonucleoprotein K/physiology , Humans , MAP Kinase Signaling System/physiology , Protein Isoforms/genetics , Protein Isoforms/physiology , Protein-Tyrosine Kinases/analysis , Protein-Tyrosine Kinases/genetics
4.
Oncogene ; 31(22): 2750-60, 2012 May 31.
Article in English | MEDLINE | ID: mdl-21963845

ABSTRACT

Aberrant activation of the Wnt signaling pathway is causally involved in the formation of most colorectal cancers (CRCs). Although detailed knowledge exists regarding Wnt-regulated protein-coding genes, much less is known about the possible involvement of non-coding RNAs. Here we used TaqMan Array MicroRNA Cards, capable of detecting 664 unique human microRNAs (miRNAs), to describe changes of the miRNA transcriptome following disruption of beta-catenin/TCF4 activity in DLD1 CRC cells. Most miRNAs appeared to respond independent of host gene regulation and proximal TCF4 chromatin occupancy as inferred from expression microarray and ChIP-chip data. A module of miRNAs induced by abrogated Wnt signaling in vitro was downregulated in two independent series of human primary CRCs (n=76) relative to normal adjacent mucosa (n=34). Several of these miRNAs (miR-145, miR-126, miR-30e-3p and miR-139-5p) markedly inhibited CRC cell growth in vitro when ectopically expressed. By using an integrative approach of proteomics and expression microarrays, we found numerous mRNAs and proteins to be affected by ectopic miR-30e-3p levels. This included HELZ and PIK3C2A that were directly repressed by several miRNA binding sites as confirmed by luciferase reporter assays in combination with mutational analyses. Finally, small interfering RNA-mediated downregulation of PIK3C2A, but not HELZ, was sufficient on its own to restrict CRC cell growth. Collectively, our study demonstrates that multiple miRNAs are upregulated as a consequence of forced attenuation of Wnt signaling in CRC cells, and some of these miRNAs inhibit cell growth with concomitant suppression of several growth-stimulatory cancer-related genes.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , MicroRNAs/genetics , Oncogenes/physiology , Transcription Factors/metabolism , Transcriptome , beta Catenin/metabolism , Aged , Aged, 80 and over , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Cell Line, Tumor , Cell Proliferation , Chromatography, Liquid , Colon/metabolism , Colorectal Neoplasms/metabolism , Female , Gene Expression Profiling , Genes, Dominant , Humans , Luciferases/metabolism , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , RNA, Small Interfering , Rectum/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Transcription Factor 4 , Transcription Factors/genetics , Tumor Cells, Cultured , Wnt Signaling Pathway , beta Catenin/genetics
5.
Oncogene ; 30(17): 2037-43, 2011 Apr 28.
Article in English | MEDLINE | ID: mdl-21242977

ABSTRACT

There is now evidence for both increased and decreased activity of the enzymes controlling the methylation of lysine 27 on histone 3 (H3K27) in cancer. One of these enzymes, KDM6B formally known as JMJD3, a histone demethylase, which removes the trimethyl mark from H3K27, is required for the lineage commitment and terminal differentiation of neural stem cells and of keratinocytes. Our results suggest that KDM6B may also have a role in antigen-driven B-cell differentiation. KDM6B expression increases in B-cell subsets with increasing stage of differentiation, and gene expression profiling shows that KDM6B transcriptional targets in germinal centre B (GC B) cells are significantly enriched for those differentially expressed during memory and plasma cell differentiation. Our results also suggest that aberrant expression of KDM6B may contribute to the pathogenesis of Hodgkin's Lymphoma (HL), an Epstein-Barr virus (EBV) associated malignancy. KDM6B is over-expressed in primary HL and induced by the EBV oncogene, latent membrane protein (LMP1) in GC B cells, the presumptive progenitors of HL. Consistent with these observations, we found that KDM6B transcriptional targets in GC B cells are enriched for genes differentially expressed in HL, and that KDM6B depletion can restore the tri-methylation of H3K27 on these genes.


Subject(s)
Gene Expression Regulation, Neoplastic , Herpesvirus 4, Human/physiology , Hodgkin Disease/genetics , Hodgkin Disease/virology , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/metabolism , Cell Differentiation , Cell Line, Tumor , Cell Transformation, Viral/genetics , Herpesvirus 4, Human/genetics , Hodgkin Disease/enzymology , Hodgkin Disease/pathology , Humans , Transcription, Genetic , Up-Regulation , Viral Matrix Proteins/genetics
6.
Article in English | MEDLINE | ID: mdl-19022750

ABSTRACT

The generation of different cell types from stem cells containing identical genetic information and their organization into tissues and organs during development is a highly complex process that requires defined transcriptional programs. Maintenance of such programs is epigenetically regulated and the factors involved in these processes are often essential for development. The activities required for cell-fate decisions are frequently deregulated in human tumors, and the elucidation of the molecular mechanisms that regulate these processes is therefore important for understanding both developmental processes and tumorigenesis.


Subject(s)
Cell Differentiation/genetics , Cell Differentiation/physiology , Histone-Lysine N-Methyltransferase/metabolism , Oxidoreductases, N-Demethylating/metabolism , Stem Cells/cytology , Stem Cells/enzymology , Animals , DNA Methylation , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/genetics , Histones/metabolism , Humans , Models, Biological , Oxidoreductases, N-Demethylating/genetics , Polycomb-Group Proteins , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription, Genetic
7.
Oncogene ; 27(43): 5706-16, 2008 Sep 25.
Article in English | MEDLINE | ID: mdl-18521079

ABSTRACT

Death-associated protein kinase 2 (DAPK2) belongs to a family of proapoptotic Ca(2+)/calmodulin-regulated serine/threonine kinases. We recently identified DAPK2 as an enhancing factor during granulocytic differentiation. To identify transcriptional DAPK2 regulators, we cloned 2.7 kb of the 5'-flanking region of the DAPK2 gene. We found that E2F1 and Krüppel-like factor 6 (KLF6) strongly activate the DAPK2 promoter. We mapped the E2F1 and KLF6 responsive elements to a GC-rich region 5' of exon 1 containing several binding sites for KLF6 and Sp1 but not for E2F. Moreover, we showed that transcriptional activation of DAPK2 by E2F1 and KLF6 is dependent on Sp1 using Sp1/KLF6-deficient insect cells, mithramycin A treatment to block Sp1-binding or Sp1 knockdown cells. Chromatin immunoprecipitation revealed recruitment of Sp1 and to lesser extent that of E2F1 and KLF6 to the DAPK2 promoter. Activation of E2F1 in osteosarcoma cells led to an increase of endogenous DAPK2 paralleled by cell death. Inhibition of DAPK2 expression resulted in significantly reduced cell death upon E2F1 activation. Similarly, KLF6 expression in H1299 cells increased DAPK2 levels accompanied by cell death that is markedly decreased upon DAPK2 knockdown. Moreover, E2F1 and KLF6 show cooperation in activating the DAPK2 promoter. In summary, our findings establish DAPK2 as a novel Sp1-dependent target gene for E2F1 and KLF6 in cell death response.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Apoptosis , Calcium-Calmodulin-Dependent Protein Kinases/genetics , E2F1 Transcription Factor/physiology , Kruppel-Like Transcription Factors/physiology , Proto-Oncogene Proteins/physiology , Apoptosis Regulatory Proteins/physiology , Binding Sites , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Cells, Cultured , Death-Associated Protein Kinases , Humans , Kruppel-Like Factor 6 , Promoter Regions, Genetic , Sp1 Transcription Factor/physiology
8.
Oncogene ; 25(2): 230-9, 2006 Jan 12.
Article in English | MEDLINE | ID: mdl-16158053

ABSTRACT

ARHI is a maternally imprinted tumor suppressor gene whose expression is markedly downregulated in breast cancer. Reactivation of ARHI expression in breast cancer cells is associated with increased histone H3 acetylation and decreased lysine 9 methylation of histone H3. An ARHI promoter segment that spanned bases -420 to +58 (designated the P2 region) exhibits significantly higher promoter activity in normal cells than in cancer cells. To better understand the molecular mechanisms contributing to this differential transcriptional activity, we sought to identify transcription factors that bind to the P2 region of the ARHI promoter and regulate its activity. Sequence analysis and oligonucleotide competition in electrophoretic mobility shift assays identified an A2 fragment containing an E2F-binding site. Using specific antibodies in supershift assays, we have shown that anti-E2F1 and 4 antibodies can supershift the A2-protein complexes, whereas anti-E2F2 and 6 antibodies cannot, demonstrating that the A2 fragment interacts with specific members of the E2F family proteins. When compared with normal breast epithelial cells, breast cancer cells have significantly elevated expression of E2F1, 4 and increased E2F DNA-binding activity. Moreover, chromatin immunoprecipitation experiments revealed that both E2F1 and 4 bind to the ARHI promoter in breast cancer cells in vivo. This binding was reduced when the cells were treated with the histone deacetylase (HDAC) inhibitor--trichostatin A (TSA). When SKBr3 cells were cotransfected with an ARHI/luciferase reporter and E2F-expression vectors, E2F1 and 4 reduced ARHI promoter activity 2-3-fold, and this reduction could be reversed by TSA treatment. The negative regulation by E2F-HDAC complexes could also be reduced by small interfering RNA of E2F1 and 4. While the retinoblastoma protein, pRB, alone had no effect on ARHI promoter activity, repression by E2F1, but not E2F4, was enhanced by the coexpression of pRB. Taken together, our results suggest that E2F1, 4 and their complexes with HDAC play an important role in downregulating the expression of the tumor suppressor gene ARHI in breast cancer cells.


Subject(s)
Breast Neoplasms/genetics , E2F1 Transcription Factor/metabolism , E2F4 Transcription Factor/metabolism , Gene Expression Regulation/genetics , Histone Deacetylases/metabolism , rho GTP-Binding Proteins/genetics , Acetylation , Binding Sites , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Nucleus/metabolism , Cells, Cultured , Chromatin Immunoprecipitation , E2F1 Transcription Factor/antagonists & inhibitors , E2F1 Transcription Factor/genetics , E2F2 Transcription Factor/antagonists & inhibitors , E2F2 Transcription Factor/genetics , E2F2 Transcription Factor/metabolism , E2F4 Transcription Factor/antagonists & inhibitors , E2F4 Transcription Factor/genetics , E2F6 Transcription Factor/antagonists & inhibitors , E2F6 Transcription Factor/genetics , E2F6 Transcription Factor/metabolism , Electrophoretic Mobility Shift Assay , Enzyme Inhibitors/pharmacology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Genes, Tumor Suppressor , Histone Deacetylase Inhibitors , Histones/metabolism , Humans , Hydroxamic Acids/pharmacology , Luciferases/metabolism , Mammary Glands, Human/cytology , Mammary Glands, Human/metabolism , Osteosarcoma/genetics , Osteosarcoma/metabolism , Osteosarcoma/pathology , Promoter Regions, Genetic/genetics , RNA, Small Interfering/pharmacology , Response Elements , Retinoblastoma Protein/metabolism , rho GTP-Binding Proteins/metabolism
9.
Apoptosis ; 7(2): 167-71, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11865201

ABSTRACT

Apoptosis Protease-Activating Factor 1, APAF1, was originally isolated four years ago and shown to be the mammalian homologue of the C. elegans pro-apoptotic ced4 gene. Since then, the expression of APAF1 has been demonstrated to be involved in several cell death pathways, including the induction of apoptosis by the p53 tumour suppressor protein and neuronal apoptosis. In this review we will focus on the regulation of APAF1 expression, in particular with regard to recent developments in our understanding of the role of APAF1 in both tumourigenesis and mammalian development.


Subject(s)
Apoptosis , Gene Expression Regulation, Developmental , Neoplasms/metabolism , Proteins/genetics , Apoptotic Protease-Activating Factor 1 , Humans , Models, Biological , Proteins/metabolism , Tumor Suppressor Protein p53/metabolism
10.
EMBO J ; 20(22): 6371-82, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11707408

ABSTRACT

Mad2 is a key component of the spindle checkpoint, a device that controls the fidelity of chromosome segregation in mitosis. The ability of Mad2 to form oligomers in vitro has been correlated with its ability to block the cell cycle upon injection into Xenopus embryos. Here we show that Mad2 forms incompatible complexes with Mad1 and Cdc20, neither of which requires Mad2 oligomerization. A monomeric point mutant of Mad2 can sustain a cell cycle arrest of comparable strength to that of the wild-type protein. We show that the interaction of Mad2 with Mad1 is crucial for the localization of Mad2 to kinetochores, where Mad2 interacts with Cdc20. We propose a model that features the kinetochore as a 'folding factory' for the formation of a Mad2-Cdc20 complex endowed with inhibitory activity on the anaphase promoting complex.


Subject(s)
Calcium-Binding Proteins/metabolism , Carrier Proteins , Cell Cycle Proteins/metabolism , Fungal Proteins/metabolism , Phosphoproteins/metabolism , Repressor Proteins/metabolism , Saccharomyces cerevisiae Proteins , Anaphase , Animals , Antibodies, Monoclonal/metabolism , Binding Sites , Cdc20 Proteins , Cell Cycle , Chromatography, Gel , DNA, Complementary/metabolism , Electrophoresis, Polyacrylamide Gel , Escherichia coli/metabolism , HeLa Cells , Humans , Kinetochores/metabolism , Mad2 Proteins , Mice , Microscopy, Fluorescence , Mitosis , Models, Biological , Mutagenesis, Site-Directed , Nuclear Proteins , Peptides/chemistry , Plasmids/metabolism , Point Mutation , Polymerase Chain Reaction , Precipitin Tests , Protein Binding , Recombinant Proteins/metabolism , Sodium Chloride/pharmacology , Transfection , Urea/pharmacology
11.
J Cell Biol ; 155(2): 207-16, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11591730

ABSTRACT

p53 is a transcriptional activator which has been implicated as a key regulator of neuronal cell death after acute injury. We have shown previously that p53-mediated neuronal cell death involves a Bax-dependent activation of caspase 3; however, the transcriptional targets involved in the regulation of this process have not been identified. In the present study, we demonstrate that p53 directly upregulates Apaf1 transcription as a critical step in the induction of neuronal cell death. Using DNA microarray analysis of total RNA isolated from neurons undergoing p53-induced apoptosis a 5-6-fold upregulation of Apaf1 mRNA was detected. Induction of neuronal cell death by camptothecin, a DNA-damaging agent that functions through a p53-dependent mechanism, resulted in increased Apaf1 mRNA in p53-positive, but not p53-deficient neurons. In both in vitro and in vivo neuronal cell death processes of p53-induced cell death, Apaf1 protein levels were increased. We addressed whether p53 directly regulates Apaf1 transcription via the two p53 consensus binding sites in the Apaf1 promoter. Electrophoretic mobility shift assays demonstrated p53-DNA binding activity at both p53 consensus binding sequences in extracts obtained from neurons undergoing p53-induced cell death, but not in healthy control cultures or when p53 or the p53 binding sites were inactivated by mutation. In transient transfections in a neuronal cell line with p53 and Apaf1 promoter-luciferase constructs, p53 directly activated the Apaf1 promoter via both p53 sites. The importance of Apaf1 as a p53 target gene in neuronal cell death was evaluated by examining p53-induced apoptotic pathways in primary cultures of Apaf1-deficient neurons. Neurons treated with camptothecin were significantly protected in the absence of Apaf1 relative to those derived from wild-type littermates. Together, these results demonstrate that Apaf1 is a key transcriptional target for p53 that plays a pivotal role in the regulation of apoptosis after neuronal injury.


Subject(s)
Apoptosis , Neurons/metabolism , Proteins/genetics , Transcriptional Activation , Tumor Suppressor Protein p53/physiology , Animals , Apoptotic Protease-Activating Factor 1 , Base Sequence , Brain Ischemia/metabolism , Brain Ischemia/pathology , Camptothecin/pharmacology , Cell Line , Cells, Cultured , Mice , Mice, Transgenic , Neurons/pathology , Promoter Regions, Genetic , Protein Biosynthesis , Proteins/physiology , RNA, Messenger/biosynthesis
12.
Eur J Biochem ; 268(18): 4969-78, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11559366

ABSTRACT

Gene expression of the plasminogen activation system is cell-cycle dependent. Previously, we showed that ectopic expression of E2F1 repressed the plasminogen activator inhibitor type 1 (PAI-1) promoter in a manner dependent on the presence of DNA-binding and transactivation domains of E2F1 but independent of binding to pocket-binding proteins, suggesting a novel mechanism for E2F-mediated negative gene regulation [Koziczak, M., Krek, W. & Nagamine, Y. (2000) Mol. Cell. Biol. 20, 2014-2022]. However, it remains to be seen whether endogenous E2F can exert a similar effect. We report here that down-regulation of PAI-1 gene expression correlates with an increase in endogenous E2F activity. When cells were treated with a cdk2/4-specific inhibitor, which maintains E2F in an inactive state, the decline of serum-induced PAI-1 mRNA levels was suppressed. In mutant U2OS cells expressing a temperature-sensitive retinoblastoma protein (pRB), a shift to a permissive temperature induced PAI-1 mRNA expression. In U2OS cells stably expressing an E2F1-estrogen receptor chimeric protein that could be activated by tamoxifen, PAI-1 gene transcription was markedly reduced by tamoxifen even in the presence of cycloheximide. These results all indicate that endogenous E2F can directly repress the PAI-1 gene. DNase I hypersensitive-site analysis of the PAI-1 promoter suggested the involvement of conformation changes in chromatin structure of the PAI-1 promoter. 5' deletion analysis of the PAI-1 promoter showed that multiple sites were responsible for the E2F negative regulation, some of which were promoter dependent. Interestingly, one of these sites is a p53-binding element.


Subject(s)
CDC2-CDC28 Kinases , Cell Cycle Proteins , DNA-Binding Proteins , Down-Regulation , Plasminogen Activator Inhibitor 1/genetics , Proto-Oncogene Proteins , Repressor Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Animals , Binding Sites , Cell Cycle , Cell Line , Chromatin/chemistry , Chromatin/genetics , Chromatin/metabolism , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , DNA/genetics , DNA/metabolism , DNA Footprinting , Deoxyribonuclease I/metabolism , E2F Transcription Factors , E2F1 Transcription Factor , Humans , Mutation , Promoter Regions, Genetic/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/genetics , Response Elements/genetics , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Swine , Temperature , Transcription Factors/genetics , Tumor Suppressor Protein p53/metabolism , Up-Regulation
13.
Nat Cell Biol ; 3(6): 552-8, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11389439

ABSTRACT

Loss of function of the retinoblastoma protein, pRB, leads to lack of differentiation, hyperproliferation and apoptosis. Inactivation of pRB results in deregulated E2F activity, which in turn induces entry to S-phase and apoptosis. Induction of apoptosis by either the loss of pRB or the deregulation of E2F activity occurs via both p53-dependent and p53-independent mechanisms. The mechanism by which E2F induces apoptosis is still unclear. Here we show that E2F1 directly regulates the expression of Apaf-1, the gene for apoptosis protease-activating factor 1. These results provide a direct link between the deregulation of the pRB pathway and apoptosis. Furthermore, because the pRB pathway is functionally inactivated in most cancers, the identification of Apaf-1 as a transcriptional target for E2F might explain the increased sensitivity of tumour cells to chemotherapy. We also show that, independently of the pRB pathway, Apaf-1 is a direct transcriptional target of p53, suggesting that p53 might sensitize cells to apoptosis by increasing Apaf-1 levels.


Subject(s)
Cell Cycle Proteins , DNA-Binding Proteins , Promoter Regions, Genetic/genetics , Proteins/genetics , Transcription Factors/physiology , Tumor Suppressor Protein p53/physiology , Animals , Apoptotic Protease-Activating Factor 1 , E2F Transcription Factors , E2F1 Transcription Factor , Embryo, Mammalian/metabolism , Humans , Mice , Proteins/metabolism , Retinoblastoma Protein/metabolism , Transcription, Genetic , Transcriptional Activation
14.
Genes Dev ; 15(3): 267-85, 2001 Feb 01.
Article in English | MEDLINE | ID: mdl-11159908

ABSTRACT

The retinoblastoma protein (pRB) and its two relatives, p107 and p130, regulate development and cell proliferation in part by inhibiting the activity of E2F-regulated promoters. We have used high-density oligonucleotide arrays to identify genes in which expression changed in response to activation of E2F1, E2F2, and E2F3. We show that the E2Fs control the expression of several genes that are involved in cell proliferation. We also show that the E2Fs regulate a number of genes involved in apoptosis, differentiation, and development. These results provide possible genetic explanations to the variety of phenotypes observed as a consequence of a deregulated pRB/E2F pathway.


Subject(s)
Apoptosis/genetics , Carrier Proteins , Cell Cycle Proteins , Cell Differentiation/genetics , DNA-Binding Proteins , Gene Expression Regulation , Transcription Factors/physiology , Blotting, Northern , Cell Cycle/genetics , Cell Division/genetics , DNA Replication , E2F Transcription Factors , E2F1 Transcription Factor , E2F2 Transcription Factor , E2F3 Transcription Factor , Gene Expression Profiling , Gene Targeting , Humans , Oligonucleotide Array Sequence Analysis , Protein Isoforms/physiology , Retinoblastoma Protein/physiology , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1 , Tumor Cells, Cultured
15.
Cell Mol Life Sci ; 57(8-9): 1193-206, 2000 Aug.
Article in English | MEDLINE | ID: mdl-11028912

ABSTRACT

To elicit the transcriptional response following intra- or extracellular stimuli, the signals need to be transmitted to their site of action within the nucleus. The nucleocytoplasmic shuttling of transcription factors is a mechanism mediating this process. The activation and inactivation of the transcriptional response is essential for cells to progress through the cell cycle in a normal manner. The involvement of cytoplasmic and nuclear accessory molecules, and the general nuclear membrane transport components, are essential for this process. Although nuclear import and export for different transcription factor families are regulated by similar mechanisms, there are several differences that allow for the specific activation of each transcription factor. This review discusses the general import and export pathways found to be common amongst many different transcription factors, and highlights a select group of transcription factors that demonstrate the diversity displayed in their mode of activation and inactivation.


Subject(s)
Cell Nucleus/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Animals , Cell Cycle , Cytoplasm/metabolism , Gene Expression Regulation , Humans , Protein Transport
16.
Mol Cell Biol ; 20(19): 7059-67, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10982822

ABSTRACT

Expression of the bovine papillomavirus E2 protein in cervical carcinoma cells represses expression of integrated human papillomavirus (HPV) E6/E7 oncogenes, followed by repression of the cdc25A gene and other cellular genes required for cell cycle progression, resulting in dramatic growth arrest. To explore the mechanism of repression of cell cycle genes in cervical carcinoma cells following E6/E7 repression, we analyzed regulation of the cdc25A promoter, which contains two consensus E2F binding sites and a consensus E2 binding site. The wild-type E2 protein inhibited expression of a luciferase gene linked to the cdc25A promoter in HT-3 cervical carcinoma cells. Mutation of the distal E2F binding site in the cdc25A promoter abolished E2-induced repression, whereas mutation of the proximal E2F site or the E2 site had no effect. None of these mutations affected the activity of the promoter in the absence of E2 expression. Expression of the E2 protein also led to posttranscriptional increase in the level of E2F4, p105(Rb), and p130 and induced the formation of nuclear E2F4-p130 and E2F4-p105(Rb) complexes. This resulted in marked rearrangement of the protein complexes that formed at the distal E2F site in the cdc25A promoter, including the replacement of free E2F complexes with E2F4-p105(Rb) complexes. These experiments indicated that repression of E2F-responsive promoters following HPV E6/E7 repression was mediated by activation of the Rb tumor suppressor pathway and the assembly of repressing E2F4-Rb DNA binding complexes. Importantly, these experiments revealed that HPV-induced alterations in E2F transcription complexes that occur during cervical carcinogenesis are reversed by repression of HPV E6/E7 expression.


Subject(s)
Carcinoma, Squamous Cell/pathology , Carrier Proteins , Cell Cycle Proteins , Gene Expression Regulation, Neoplastic , Gene Expression Regulation, Viral , Neoplasm Proteins/metabolism , Papillomaviridae/genetics , Proteins , Retinoblastoma Protein/metabolism , Transcription Factors/metabolism , Uterine Cervical Neoplasms/pathology , Viral Proteins/biosynthesis , Viral Proteins/physiology , cdc25 Phosphatases/genetics , Binding Sites , Bovine papillomavirus 1/genetics , Bovine papillomavirus 1/physiology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/virology , Cell Cycle/genetics , Cell Cycle/physiology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Viral/genetics , Consensus Sequence , Cysteine Endopeptidases/metabolism , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , E2F Transcription Factors , E2F4 Transcription Factor , Female , Genes, Retinoblastoma , Humans , Macromolecular Substances , Multienzyme Complexes/metabolism , Neoplasm Proteins/genetics , Papillomaviridae/physiology , Papillomavirus Infections/genetics , Papillomavirus Infections/metabolism , Papillomavirus Infections/pathology , Papillomavirus Infections/virology , Phosphoproteins/biosynthesis , Phosphoproteins/genetics , Promoter Regions, Genetic , Proteasome Endopeptidase Complex , Protein Binding , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Retinoblastoma Protein/biosynthesis , Retinoblastoma Protein/genetics , Retinoblastoma-Binding Protein 1 , Retinoblastoma-Like Protein p130 , Transcription Factor DP1 , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transfection , Tumor Cells, Cultured/metabolism , Tumor Virus Infections/genetics , Tumor Virus Infections/metabolism , Tumor Virus Infections/pathology , Tumor Virus Infections/virology , Uterine Cervical Neoplasms/genetics
17.
Genes Dev ; 14(18): 2330-43, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-10995389

ABSTRACT

CDC6 is conserved during evolution and is essential and limiting for the initiation of eukaryotic DNA replication. Human CDC6 activity is regulated by periodic transcription and CDK-regulated subcellular localization. Here, we show that, in addition to being absent from nonproliferating cells, CDC6 is targeted for ubiquitin-mediated proteolysis by the anaphase promoting complex (APC)/cyclosome in G(1). A combination of point mutations in the destruction box and KEN-box motifs in CDC6 stabilizes the protein in G(1) and in quiescent cells. Furthermore, APC, in association with CDH1, ubiquitinates CDC6 in vitro, and both APC and CDH1 are required and limiting for CDC6 proteolysis in vivo. Although a stable mutant of CDC6 is biologically active, overexpression of this mutant or wild-type CDC6 is not sufficient to induce multiple rounds of DNA replication in the same cell cycle. The APC-CDH1-dependent proteolysis of CDC6 in early G(1) and in quiescent cells suggests that this process is part of a mechanism that ensures the timely licensing of replication origins during G(1).


Subject(s)
Cell Cycle Proteins/metabolism , DNA Replication , G1 Phase , Ligases/metabolism , Saccharomyces cerevisiae Proteins , Ubiquitin-Protein Ligase Complexes , Amino Acid Sequence , Anaphase-Promoting Complex-Cyclosome , Animals , Blotting, Northern , Blotting, Western , Cell Cycle , Cell Cycle Proteins/genetics , Cell Division , Cell Line , Cysteine Endopeptidases/metabolism , Fluorescent Antibody Technique , Humans , Molecular Sequence Data , Multienzyme Complexes/metabolism , Phosphorylation , Point Mutation , Proteasome Endopeptidase Complex , RNA, Messenger/metabolism , Replication Origin , Ubiquitin-Protein Ligases , Ubiquitins/metabolism
18.
Clin Cancer Res ; 6(4): 1488-97, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10778981

ABSTRACT

The transcription factor complex E2F-1/DP-1 regulates the G1-to-S-phase transition and has been associated with sensitivity to the S-phase-specific anticancer agents camptothecin and etoposide, which poison DNA topoisomerase I and II, respectively. To investigate the relationship between E2F-1 and drug sensitivity in detail, we established human osteosarcoma U-20S-TA cells expressing full-length E2F-1/ DP-1 under the control of a tetracycline-responsive promoter, designated UE1DP-1 cells. Topoisomerase I levels and activity as well as the number of camptothecin-induced DNA single- and double-strand breaks were unchanged in UEIDP-1/tc- cells with >10-fold E2F-1/DP-1 overexpression. However, UE1DP-1/tc- cells were hypersensitive to camptothecin in both a clonogenic assay and four different apoptotic assays. This indicates that camptothecin-induced toxicity in this model is due to the activation of an E2F-1/ DP-1-induced post-DNA damage pathway rather than an increase in the number of replication forks caused by the S-phase initiation. In contrast, topoisomerase IIalpha levels (but not topoisomerase IIbeta levels), together with topoisomerase IIalpha promoter activity, increased 2--3-fold in UE1DP-1/tc-cells. Furthermore, the number of etoposide-induced DNA single- and double-strand breaks increased in UE1DP-1/tc-cells together with a rise in clonogenic sensitivity to etoposide, but an equal apoptotic sensitivity to etoposide. The increase in topoisomerase IIalpha promoter activity in UE1DP-1/tc--cells was shown to be due to S-phase initiation per se because it was blocked by ectopic expression of dominant negative cyclin-dependent kinase 2. In conclusion, overexpression of E2F-1/DP-1 in U-20S-TA cells is sufficient to increase clonogenic sensitivity to both topoisomerase I- and II-targeted anticancer drugs. However, the mechanism by which this occurs appears to be qualitatively different. The UE1DP-1 cell model may be used to elucidate post-DNA damage mechanisms of cell death induced by topoisomerase I-directed anticancer agents.


Subject(s)
Antineoplastic Agents/pharmacology , Carrier Proteins , Cell Cycle Proteins , Topoisomerase I Inhibitors , Topoisomerase II Inhibitors , Transcription Factors/metabolism , Apoptosis/drug effects , Camptothecin/pharmacology , Cell Cycle , Cell Survival/drug effects , Cisplatin/pharmacology , DNA/drug effects , DNA/genetics , DNA/metabolism , DNA Damage/drug effects , DNA Topoisomerases, Type I/metabolism , DNA Topoisomerases, Type II/genetics , DNA Topoisomerases, Type II/metabolism , DNA, Recombinant/genetics , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/metabolism , E2F Transcription Factors , E2F1 Transcription Factor , Enzyme Inhibitors/pharmacology , Etoposide/pharmacology , Gene Expression Regulation, Neoplastic , Humans , Luciferases/genetics , Luciferases/metabolism , Promoter Regions, Genetic/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Retinoblastoma-Binding Protein 1 , S Phase , Thymidine/metabolism , Transcription Factor DP1 , Transcription Factors/genetics , Tumor Cells, Cultured
19.
J Biol Chem ; 275(25): 19258-67, 2000 Jun 23.
Article in English | MEDLINE | ID: mdl-10748129

ABSTRACT

In animal cells the progression of the cell cycle through G(1)/S transition and S phase is under the control of the pRB/E2F regulatory pathway. The E2F transcription factors are key activators of genes coding for several regulatory proteins and for enzymes involved in nucleotide and DNA synthesis. In this report we have detected the presence of E2F-like DNA binding activities in carrot nuclear extracts, and we have isolated a carrot cDNA (DcE2F) encoding a plant E2F homologue. The DcE2F gene is expressed in proliferating cells and is induced during the G(1)/S transition of the cell cycle. Supershift experiments using anti-DcE2F antiserum have confirmed that the DcE2F protein is a component of the carrot E2F-like nuclear activities. DNA binding assays have demonstrated that the DcE2F protein can recognize a canonical E2F cis-element in association with a mammalian DP protein. Furthermore, transactivation assays have revealed that DcE2F is a functional transcription factor that can transactivate, together with a DP partner, an E2F-responsive reporter gene in both plant and mammalian cells.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , DNA-Binding Proteins , Daucus carota/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cell Cycle , DNA, Complementary , Daucus carota/cytology , E2F Transcription Factors , Molecular Sequence Data , Retinoblastoma-Binding Protein 1 , Sequence Homology, Amino Acid , Trans-Activators/chemistry , Trans-Activators/genetics , Transcription Factors/chemistry , Transcription Factors/genetics
20.
Biochim Biophys Acta ; 1470(1): M1-12, 2000 Feb 14.
Article in English | MEDLINE | ID: mdl-10656985

ABSTRACT

Ever since its discovery, the RB-1 gene and the corresponding protein, pRB, have been a focal point of cancer research. The isolation of E2F transcription factors provided the key to our current understanding of RB-1 function in the regulation of the cell cycle and in tumor suppression. It is becoming more and more evident that the regulatory circuits governing the cell cycle are very complex and highly interlinked. Certain aspects of RB-1 function, for instance its role in differentiation, cannot be easily explained by the current models of pRB-E2F interaction. One reason is that pRB has targets different from E2F, molecules like MyoD for instance. Another reason may be that we have not completely understood the full complexity of E2F function, itself. In this review, we will try to illuminate the role of E2F in pRB- and p53-mediated tumor suppression pathways with particular emphasis on the aspect of E2F-mediated transcriptional regulation. We conclude that E2F can mediate transcriptional activation as well as transcriptional repression of E2F target genes. The net effect of E2F on the transcriptional activity of a particular gene may be the result of as yet poorly understood protein-protein interactions of E2F with other components of the transcriptional machinery, as well as it may reflect the readout of the different ways of regulating E2F activity, itself. We will discuss the relevance of a thorough understanding of E2F function for cancer therapy.


Subject(s)
Carrier Proteins , Cell Cycle Proteins/genetics , Cell Division/genetics , DNA-Binding Proteins , Transcription Factors/genetics , Animals , Apoptosis , E2F Transcription Factors , Genes, Tumor Suppressor , Humans , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1 , Transcriptional Activation , Tumor Suppressor Protein p53/genetics
SELECTION OF CITATIONS
SEARCH DETAIL