Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Genes Dev ; 29(17): 1875-89, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26314710

ABSTRACT

The retinoblastoma tumor suppressor (pRb) protein associates with chromatin and regulates gene expression. Numerous studies have identified Rb-dependent RNA signatures, but the proteomic effects of Rb loss are largely unexplored. We acutely ablated Rb in adult mice and conducted a quantitative analysis of RNA and proteomic changes in the colon and lungs, where Rb(KO) was sufficient or insufficient to induce ectopic proliferation, respectively. As expected, Rb(KO) caused similar increases in classic pRb/E2F-regulated transcripts in both tissues, but, unexpectedly, their protein products increased only in the colon, consistent with its increased proliferative index. Thus, these protein changes induced by Rb loss are coupled with proliferation but uncoupled from transcription. The proteomic changes in common between Rb(KO) tissues showed a striking decrease in proteins with mitochondrial functions. Accordingly, RB1 inactivation in human cells decreased both mitochondrial mass and oxidative phosphorylation (OXPHOS) function. RB(KO) cells showed decreased mitochondrial respiratory capacity and the accumulation of hypopolarized mitochondria. Additionally, RB/Rb loss altered mitochondrial pyruvate oxidation from (13)C-glucose through the TCA cycle in mouse tissues and cultured cells. Consequently, RB(KO) cells have an enhanced sensitivity to mitochondrial stress conditions. In summary, proteomic analyses provide a new perspective on Rb/RB1 mutation, highlighting the importance of pRb for mitochondrial function and suggesting vulnerabilities for treatment.


Subject(s)
Mitochondria/metabolism , Oxidative Phosphorylation , Retinoblastoma Protein/genetics , Animals , Cells, Cultured , Colon/physiopathology , Gene Expression Regulation , Gene Knockout Techniques , Humans , Lung/physiopathology , Mice , Mitochondria/genetics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Proteomics , Retinoblastoma Protein/metabolism , Stress, Physiological/genetics , Transcriptome
2.
Genes Dev ; 27(2): 182-96, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23322302

ABSTRACT

Inactivation of the retinoblastoma tumor suppressor (pRB) alters the expression of a myriad of genes. To understand the altered cellular environment that these changes create, we took advantage of the Drosophila model system and used targeted liquid chromatography tandem mass spectrometry (LC-MS/MS) to profile the metabolic changes that occur when RBF1, the fly ortholog of pRB, is removed. We show that RBF1-depleted tissues and larvae are sensitive to fasting. Depletion of RBF1 causes major changes in nucleotide synthesis and glutathione metabolism. Under fasting conditions, these changes interconnect, and the increased replication demand of RBF1-depleted larvae is associated with the depletion of glutathione pools. In vivo (13)C isotopic tracer analysis shows that RBF1-depleted larvae increase the flux of glutamine toward glutathione synthesis, presumably to minimize oxidative stress. Concordantly, H(2)O(2) preferentially promoted apoptosis in RBF1-depleted tissues, and the sensitivity of RBF1-depleted animals to fasting was specifically suppressed by either a glutamine supplement or the antioxidant N-acetyl-cysteine. Effects of pRB activation/inactivation on glutamine catabolism were also detected in human cell lines. These results show that the inactivation of RB proteins causes metabolic reprogramming and that these consequences of RBF/RB function are present in both flies and human cell lines.


Subject(s)
Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Glutamine/biosynthesis , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , Apoptosis , Cell Line, Tumor , DNA Damage , Fasting/metabolism , Glutathione/biosynthesis , Humans , Larva , Mutation , Nucleotides/biosynthesis , Oxidative Stress , Retinoblastoma Protein , Stress, Physiological
3.
Genes Dev ; 26(4): 356-68, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22345517

ABSTRACT

E2F transcription factors are important regulators of cell proliferation and are frequently dysregulated in human malignancies. To identify novel regulators of E2F function, we used Drosophila as a model system to screen for mutations that modify phenotypes caused by reduced levels of dE2F1. This screen identified components of the Pumilio translational repressor complex (Pumilio, Nanos, and Brain tumor) as suppressors of dE2F1-RNAi phenotypes. Subsequent experiments provided evidence that Pumilio complexes repress dE2F1 levels and that this mechanism of post-transcriptional regulation is conserved in human cells. The human Pumilio homologs Pum 1 and Pum 2 repress the translation of E2F3 by binding to the E2F3 3' untranslated region (UTR) and also enhance the activity of multiple E2F3 targeting microRNAs (miRNAs). E2F3 is an oncogene with strong proliferative potential and is regularly dysregulated or overexpressed in cancer. Interestingly, Pumilio/miRNA-mediated regulation of E2F3 is circumvented in cancer cells in several different ways. Bladder carcinomas selectively down-regulate miRNAs that cooperate with Pumilio to target E2F3, and multiple tumor cell lines shorten the 3' end of the E2F3 mRNA, removing the Pumilio regulatory elements. These studies suggest that Pumilio-miRNA repression of E2F3 translation provides an important level of E2F regulation that is frequently abrogated in cancer cells.


Subject(s)
Drosophila Proteins/metabolism , E2F Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/metabolism , RNA-Binding Proteins/metabolism , Transcription Factors/metabolism , 3' Untranslated Regions/genetics , Animals , Cell Line, Tumor , Drosophila , Drosophila Proteins/genetics , E2F Transcription Factors/metabolism , HeLa Cells , Humans , MicroRNAs/genetics , RNA Processing, Post-Transcriptional , RNA-Binding Proteins/genetics , Transcription Factors/genetics
4.
Genes Dev ; 25(8): 814-30, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21498571

ABSTRACT

pRB-mediated inhibition of cell proliferation is a complex process that depends on the action of many proteins. However, little is known about the specific pathways that cooperate with the Retinoblastoma protein (pRB) and the variables that influence pRB's ability to arrest tumor cells. Here we describe two shRNA screens that identify kinases that are important for pRB to suppress cell proliferation and pRB-mediated induction of senescence markers. The results reveal an unexpected effect of LATS2, a component of the Hippo pathway, on pRB-induced phenotypes. Partial knockdown of LATS2 strongly suppresses some pRB-induced senescence markers. Further analysis shows that LATS2 cooperates with pRB to promote the silencing of E2F target genes, and that reduced levels of LATS2 lead to defects in the assembly of DREAM (DP, RB [retinoblastoma], E2F, and MuvB) repressor complexes at E2F-regulated promoters. Kinase assays show that LATS2 can phosphorylate DYRK1A, and that it enhances the ability of DYRK1A to phosphorylate the DREAM subunit LIN52. Intriguingly, the LATS2 locus is physically linked with RB1 on 13q, and this region frequently displays loss of heterozygosity in human cancers. Our results reveal a functional connection between the pRB and Hippo tumor suppressor pathways, and suggest that low levels of LATS2 may undermine the ability of pRB to induce a permanent cell cycle arrest in tumor cells.


Subject(s)
Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/physiology , Retinoblastoma Protein/metabolism , Tumor Suppressor Proteins/metabolism , Cell Cycle/genetics , Cell Cycle/physiology , Cell Line , Cell Line, Tumor , Cell Proliferation , Chromatin Immunoprecipitation , Flow Cytometry , Humans , Immunoblotting , Loss of Heterozygosity/genetics , Oligonucleotide Array Sequence Analysis , Phosphorylation , Polymerase Chain Reaction , Protein Binding/genetics , Protein Binding/physiology , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , RNA, Small Interfering/genetics , Retinoblastoma Protein/genetics , Signal Transduction/genetics , Signal Transduction/physiology , Tumor Suppressor Proteins/genetics , Dyrk Kinases
5.
Mol Cell ; 40(5): 736-48, 2010 Dec 10.
Article in English | MEDLINE | ID: mdl-21145482

ABSTRACT

The KDM4/JMJD2 family of histone demethylases is amplified in human cancers. However, little is known about their physiologic or tumorigenic roles. We have identified a conserved and unappreciated role for the JMJD2A/KDM4A H3K9/36 tridemethylase in cell cycle progression. We demonstrate that JMJD2A protein levels are regulated in a cell cycle-dependent manner and that JMJD2A overexpression increased chromatin accessibility, S phase progression, and altered replication timing of specific genomic loci. These phenotypes depended on JMJD2A enzymatic activity. Strikingly, depletion of the only C. elegans homolog, JMJD-2, slowed DNA replication and increased ATR/p53-dependent apoptosis. Importantly, overexpression of HP1γ antagonized JMJD2A-dependent progression through S phase, and depletion of HPL-2 rescued the DNA replication-related phenotypes in jmjd-2(-/-) animals. Our findings describe a highly conserved model whereby JMJD2A regulates DNA replication by antagonizing HP1γ and controlling chromatin accessibility.


Subject(s)
Cell Cycle , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cells, Cultured , Chromatin Immunoprecipitation , Chromosomal Proteins, Non-Histone/antagonists & inhibitors , Chromosomal Proteins, Non-Histone/genetics , DNA Replication , Flow Cytometry , HeLa Cells , Humans , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Transfection
6.
Onkologie ; 32(5): 264-72, 2009 May.
Article in English | MEDLINE | ID: mdl-19420973

ABSTRACT

BACKGROUND: In multimodal therapy concepts for advanced head and neck squamous cell carcinoma (HNSCC), a valid predictive assay for the quick detection of efficient chemotherapeutic agents is desirable. Questionable so far was whether tissue samples of about 100 mg correctly reflect the chemoresponse of a whole HNSCC. This was proven using an ex-vivo colony-forming assay. MATERIALS AND METHODS: Of 14 HNSCC, 3 biopsies each were taken from separate sites, minced, and collagenase digested. HNSCC digests were added to microtiter plates containing serial dilutions of chemotherapeutic agents or medium as control. After 72-h incubation, wells were washed and cultures methanol-fixed before Giemsa-staining. Epithelial colonies were counted. RESULTS: 11/14 HNSCC (78.6%) showed sufficient colony formation allowing reliable cut-off detection. Cut-off concentrations (complete chemotherapeutically suppressed colony formation) between 3.3 microM and >50 microM cisplatin, and 0.55 microM and 17.6 microM docetaxel were detected. Inhibition of colony formation to 50% of colonies detected in controls (IC50) was found between 0.2 microM and 17.9 microM cisplatin or 1.5 microM and 13.7 microM docetaxel. Cut-off concentrations and IC50 of the HNSCC fragments showed a strong correlation (docetaxel: r > 0.80, p < 0.005; cisplatin: r > 0.67, p < 0.044), while being only insignificantly different in the t-test for paired samples (docetaxel: p > 0.163; cisplatin: p > 0.167). CONCLUSION: In most cases, tissue samples of about 100 mg allow a representative assessment of chemoresponse of HNSCC.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Cisplatin/administration & dosage , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/pathology , Outcome Assessment, Health Care/methods , Taxoids/administration & dosage , Adult , Aged , Antineoplastic Agents/administration & dosage , Biopsy/methods , Docetaxel , Female , Humans , Male , Middle Aged , Reproducibility of Results , Sensitivity and Specificity , Treatment Outcome
7.
Gene ; 413(1-2): 67-75, 2008 Apr 30.
Article in English | MEDLINE | ID: mdl-18329191

ABSTRACT

The EGF-TM7 receptor CD97 shows different features of expression and function in muscle cells compared to hematopoetic and tumor cells. Since the molecular function and regulation of CD97 are poorly understood, this study aimed at defining its basal transcriptional regulation in smooth muscle cells (SMCs). The computational analysis of the CD97 5'-flanking region revealed that the TATA box-lacking promoter possesses several GC-rich regions as putative Sp1/Sp3 binding sites. Transfection studies with serially deleted promoter constructs demonstrated that the minimal promoter fragment resided in the -218/+45 region containing one out of five identified GC-boxes in the leiomyosarcoma cell line SK-LMS-1 and human bronchial smooth muscle cells (HbSMCs). Mutation of the most proximal GC-site in CD97 reporter gene constructs caused a significant decrease in promoter activity. Gel shift assays and chromatin immunoprecipitation revealed that Sp1 and Sp3 bound specifically to the most proximal GC-site. Furthermore, we showed that Sp1 and Sp3 over-expression activates CD97 promoter activity in HEK293 cells. Our data characterize for the first time the activity of the human CD97 promoter which is controlled by Sp1/Sp3 transcription factors in SMCs.


Subject(s)
Antigens, CD/genetics , Membrane Glycoproteins/genetics , Myocytes, Smooth Muscle/metabolism , Promoter Regions, Genetic , Sp1 Transcription Factor/metabolism , Sp3 Transcription Factor/metabolism , Base Sequence , Binding Sites/genetics , Cell Line , Cloning, Molecular , DNA/genetics , DNA/metabolism , DNA Primers/genetics , GC Rich Sequence , Genes, Reporter , Humans , Luciferases/genetics , Mutagenesis, Site-Directed , Receptors, G-Protein-Coupled , Transcriptional Activation
8.
Biochim Biophys Acta ; 1759(6): 270-80, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16934346

ABSTRACT

Here we describe an RT-PCR analysis of mono-ADP-ribosyltransferase 3 (ART3) mRNA expression in macrophages, testis, semen, tonsil, heart and skeletal muscle and the complete gene structure as obtained by sequence alignment of PCR products with a human genomic clone (GenBank accession no. AC112719). Twelve exons (ex1-12) were found to make up the coding region of the gene (one more than previously published). Two prominent classes of ART3 splice variants could be distinguished by the presence or absence of ex2 which encodes most of ART3 protein. Among the ex2-containing mRNA species, the most frequently amplified variant did not include exons 9 to 11, except in skeletal muscle, in which the major splice variant lacked ex10 only. Two different, previously not reported 5' non-translated regions (5' UTRs) were identified, demonstrating the presence of two alternative promoters that we termed palpha and pbeta. Whereas the 5'UTR originating from palpha, was split up into three exons, a single exon represented the 5' UTR of pbeta transcripts. Strikingly, in heart, skeletal muscle and tonsils the upstream promoter palpha was totally inactive and ART3 transcription appears to be driven solely by pbeta. In all other cell types tested, transcription started mainly (if not exclusively) at palpha. Thus, ART3 expression in human cells appears to be governed by a combination of differential splicing and tissue-preferential use of two alternative promoters. This specific use is evolutionary conserved as shown by analysis of the 5' UTR of the mouse ART3 mRNA.


Subject(s)
ADP Ribose Transferases/genetics , Membrane Proteins/genetics , 5' Untranslated Regions , Alternative Splicing , Amino Acid Sequence , Base Sequence , Cells, Cultured , Cloning, Molecular , DNA Primers/genetics , Exons , GPI-Linked Proteins , Gene Expression , Humans , In Vitro Techniques , Male , Molecular Sequence Data , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Tissue Distribution , Transfection
9.
FEBS Lett ; 581(6): 1166-72, 2007 Mar 20.
Article in English | MEDLINE | ID: mdl-17336302

ABSTRACT

Cks2 proteins are essential components of cyclin/cyclin-dependent kinase complexes and contribute to cell cycle control. We identify Cks2 as a transcriptional target downregulated by the tumor suppressor p53. Cks2 expression was found to be repressed by p53 both at the mRNA and the protein levels. p53 downregulates transcription from the Cks2 promoter in a dose-dependent manner and in all cell types tested. This repression appears to be independent of p53 binding to the Cks2 promoter. In contrast to p53, neither p63 nor p73 proteins can repress Cks2 transcription. Thus p53, rather than its homologues p63 and p73, may contribute to control of the first metaphase/anaphase transition of mammalian meiosis by downregulation of Cks2 expression.


Subject(s)
Carrier Proteins/genetics , Cell Cycle Proteins/genetics , DNA-Binding Proteins/physiology , Gene Expression Regulation , Membrane Proteins/physiology , Nuclear Proteins/physiology , Protein Kinases/genetics , Tumor Suppressor Protein p53/physiology , Tumor Suppressor Proteins/physiology , Anaphase , CDC2-CDC28 Kinases , Cell Line , Down-Regulation , Meiosis , Metaphase , Promoter Regions, Genetic , Tumor Protein p73
10.
FEBS J ; 274(20): 5235-49, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17868378

ABSTRACT

DNA methylation is an important mechanism involved in embryogenesis and tumor development. Changing cytosines to 5-methylcytosines in CpG dinucleotides has been found to be responsible for the inactivation of tumor suppressor genes by repressing transcription. A central cell cycle regulator whose synthesis is controlled by transcription is cyclin B. In mammalian cells, cyclin B1 and B2 proteins are well characterized and often found to be overexpressed in cancer patients. Transcription from cyclin B1 and B2 promoters during the cell cycle is dependent upon a combination of two sites named 'cell cycle-dependent element' (CDE) and 'cell cycle genes homology region' (CHR), through repression in G(0) and G(1) followed by release in G(2)/M. Here we show that the cyclin B2 promoter contains a CpG island and that 5-aza-deoxycytidine treatment leads to deregulation of cell cycle-dependent mRNA expression from this gene via a loss of repression in G(0). Furthermore, deletion of the DNA methyltransferase genes DNMT1 and DNMT3b leads to an increase in transcription of cyclin B2. Additionally, DNA methylation in vitro prevents transcriptional activation of the cyclin B2 promoter in G(2)/M. Analysis in vivo of the cyclin B2 core promoter revealed that the CDE/CHR site is partially methylated. However, quantitative in vivo analysis of the CpG-methylation level of the CDE during cell division indicates that CpG methylation is independent of the cell cycle. We conclude that DNA methylation affects cell cycle-dependent transcription of cyclin B2 but that regulation through CDE/CHR is independent of cytosine methylation.


Subject(s)
Cell Cycle/physiology , Cyclin B/metabolism , DNA Methylation , Promoter Regions, Genetic/genetics , Regulatory Sequences, Nucleic Acid/genetics , Animals , Cells, Cultured , Cyclin B/genetics , Cyclin B2 , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/physiology , Flow Cytometry , Gene Expression Regulation , HCT116 Cells , Humans , Luciferases/metabolism , Mice , NIH 3T3 Cells , Response Elements/genetics , Transcription, Genetic , Transcriptional Activation , DNA Methyltransferase 3B
11.
FEBS J ; 273(8): 1681-95, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16623705

ABSTRACT

Cyclins form complexes with cyclin-dependent kinases. By controlling activity of the enzymes, cyclins regulate progression through the cell cycle. A- and B-type cyclins were discovered due to their distinct appearance in S and G(2) phases and their rapid proteolytic destruction during mitosis. Transition from G(2) to mitosis is basically controlled by B-type cyclins. In mammals, two cyclin B proteins are well characterized, cyclin B1 and cyclin B2. Recently, a human cyclin B3 gene was described. In contrast to the expression pattern of other B-type cyclins, we find cyclin B3 mRNA expressed not only in S and G(2)/M cells but also in G(0) and G(1). Human cyclin B3 is expressed in different variants. We show that one isoform remains in the cytoplasm, whereas the other variant is translocated to the nucleus. Transport to the nucleus is dependent on three autonomous nonclassical nuclear localization signals that where previously not implicated in nuclear translocation. It had been shown that cyclin B3 coimmunoprecipitates with cdk2; but this complex does not exhibit any kinase activity. Furthermore, a degradation-resistant version of cyclin B3 can arrest cells in G(1) and G(2). Taken together with the finding that cyclin B3 mRNA is not only expressed in G(2)/M but is also detected in significant amounts in resting cells and in G(1) cells. This may suggest a dominant-negative function of human cyclin B3 in competition with activating cyclins in G(0) and the G(1) phase of the cell cycle.


Subject(s)
Cell Cycle/physiology , Cyclin B/genetics , Fibroblasts/metabolism , Nuclear Localization Signals/metabolism , RNA, Messenger/biosynthesis , Amino Acid Sequence , Blotting, Northern , Blotting, Western , Cell Culture Techniques , Cyclin B/metabolism , Flow Cytometry , Fluorescent Antibody Technique, Indirect , Humans , Immunoprecipitation , Molecular Sequence Data , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Skin/cytology
12.
Anticancer Res ; 26(3B): 2361-5, 2006.
Article in English | MEDLINE | ID: mdl-16821617

ABSTRACT

BACKGROUND: Vinorelbine has been identified as an effective cytostatic drug in head and neck squamous cell carcinoma (HNSCC). To predict the individual chemoresponse, the application of an ex vivo assay to quantify the response of HNSCC to vinorelbine is presented. PATIENTS AND METHODS: Twelve biopsies from primary HNSCC sites and five biopsies from metastatic lesions were analyzed (n = 17). The specimens were investigated ex vivo for the overall, epithelial and stromal chemoresponse to vinorelbine. RESULTS: By selective evaluation of the epithelial chemoresponse, the applied assay identified three specimens as vinorelbine-sensitive (18%), including one de novo sensitivity in a metastatic lesion of a vinorelbine-resistant hypopharyngeal carcinoma. CONCLUSION: Applying flavin-protecting culture methods and with careful correction for the stromal cell effect, the assay generated reliable data for the assessment of the individual chemoresponse of HNSCC specimens to vinorelbine. The assay may, therefore, facilitate the implementation of individualized chemotherapy protocols.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Carcinoma, Squamous Cell/drug therapy , Head and Neck Neoplasms/drug therapy , Vinblastine/analogs & derivatives , Carcinoma, Squamous Cell/pathology , Drug Screening Assays, Antitumor , Epithelial Cells/drug effects , Head and Neck Neoplasms/pathology , Humans , KB Cells , Neoplasm Staging , Vinblastine/pharmacology , Vinorelbine
13.
Oncogene ; 23(19): 3376-84, 2004 Apr 22.
Article in English | MEDLINE | ID: mdl-14990988

ABSTRACT

T-cell factor (Tcf)-4 is a main transcription factor to pass on Wnt/beta-catenin signalling. The tumour suppressor protein p53 contributes as a transcription factor to cell-cycle arrest and apoptosis induction. Mutations of components in p53 and Wnt/beta-catenin signalling networks play a part in tumour formation. Here, we identify the Tcf-4 gene as a downstream effector of p53. Induction of wild-type p53 in a tet-off regulated human colon cell system leads to the reduction of Tcf-4 mRNA and protein levels. Also, mRNA of the Tcf-4 target gene uPAR is downregulated after p53 induction. Expression of a luciferase reporter controlled by the Tcf-4 promoter is repressed by wild-type p53, but not by a p53 mutant deficient in DNA binding. Such a regulation is seen in cell lines of different origin. These findings directly link Wnt/beta-catenin signalling and p53 tumour suppressor function and may provide a mechanism by which loss of p53 function contributes to progression in the adenoma/carcinoma sequence in colon tumours. Furthermore, since Tcf-4 is expressed in many tissues and downregulation of Tcf-4 by p53 is seen in several different cell types, this regulation likely plays a role in proliferation control of all tissues that can express p53 and Tcf-4.


Subject(s)
Signal Transduction , Transcription Factors/genetics , Transcription, Genetic , Tumor Suppressor Protein p53/physiology , Animals , Cytoskeletal Proteins/physiology , Down-Regulation , Gene Expression Regulation , Humans , Mice , NIH 3T3 Cells , Promoter Regions, Genetic , RNA, Messenger/analysis , TCF Transcription Factors , Trans-Activators/physiology , Transcription Factor 7-Like 2 Protein , Transcription Factors/analysis , beta Catenin
14.
Gene ; 312: 225-37, 2003 Jul 17.
Article in English | MEDLINE | ID: mdl-12909359

ABSTRACT

Cyclins are essential regulators of the cell division cycle. Cyclin B associates with the cyclin-dependent kinase 1 (cdc2) to form a complex which is required for cells to undergo mitosis. In mammalian cells three B-type cyclins have been characterised, cyclin B1, B2 and B3. The cell cycle-dependent synthesis of cyclin B1 and B2 has been investigated in detail displaying maximum expression in G2 which is mainly regulated on the transcriptional level. We have previously shown that this regulation of the mouse cyclin B2 promoter is controlled by a cell cycle-dependent element (CDE) and the cell cycle genes homology region (CHR). Also in a number of other genes CDE/CHR elements repress transcription in G0 and G1 and lead to relief of repression later during the cell cycle. Here, we compare human and mouse cyclin B2 promoters. Both promoters share only nine regions with nucleotide identities. Three of these sites are CCAAT-boxes spaced 33 bp apart which can bind the NF-Y transcriptional activator. NF-Y binding to the human cyclin B2 promoter could be shown by chromatin immunoprecipitation (ChIP) assays. Activation by NF-Y is responsible for more than 93% of the total promoter activity as measured by cotransfecting a plasmid coding for a dominant-negative form of NF-YA. Cell cycle-dependent repression is regulated solely through a CHR. Surprisingly, in contrast to the mouse promoter the CHR in the human cyclin B2 promoter does not rely on a CDE site in tandem with it. Together with the recently described mouse cdc25C promoter, human cyclin B2 is the second identified gene which solely requires a CHR for its cell cycle regulation.


Subject(s)
CCAAT-Binding Factor/metabolism , Cell Cycle Proteins/genetics , Cyclin B/genetics , Promoter Regions, Genetic/genetics , Regulatory Sequences, Nucleic Acid/genetics , 3T3 Cells , Animals , Base Sequence , Binding Sites/genetics , CCAAT-Binding Factor/genetics , Cell Cycle/physiology , Cells, Cultured , Cloning, Molecular , Cyclin B2 , DNA/chemistry , DNA/genetics , Flow Cytometry/methods , Gene Expression , Humans , Luciferases/genetics , Luciferases/metabolism , Male , Mice , Molecular Sequence Data , Protein Binding , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Response Elements/genetics , Sequence Analysis, DNA , Sequence Homology, Nucleic Acid , Transcription, Genetic , Tumor Cells, Cultured
15.
FEBS Lett ; 536(1-3): 66-70, 2003 Feb 11.
Article in English | MEDLINE | ID: mdl-12586340

ABSTRACT

Cyclin B is a central regulator of transition from the G(2) phase of the cell cycle to mitosis. In mammalian cells two B-type cyclins have been characterised, cyclin B1 and B2. Both are expressed with a maximum in G(2) and their synthesis is mainly regulated on the transcriptional level. We show that a single cell cycle genes homology region, lacking a functional cell cycle-dependent element in tandem with it, contributes most of the cell cycle-dependent transcription from the cyclin B1 promoter. The coactivator p300 binds to the cyclin B1 promoter and synergises with the transcription factor NF-Y in activating transcription of cyclin B1.


Subject(s)
Cyclin B/genetics , Nuclear Proteins/physiology , Response Elements , Trans-Activators/physiology , Transcriptional Activation , 3T3 Cells , Animals , Base Sequence , Binding Sites , Cell Cycle , Cell Line , Cells, Cultured , Conserved Sequence , Cyclin B/biosynthesis , Cyclin B1 , E1A-Associated p300 Protein , Gene Expression Regulation , Humans , Kinetics , Mice , Molecular Sequence Data , Promoter Regions, Genetic , Regulatory Sequences, Nucleic Acid , Sequence Alignment
16.
Cancer Chemother Pharmacol ; 65(6): 1153-63, 2010 May.
Article in English | MEDLINE | ID: mdl-19771432

ABSTRACT

BACKGROUND: Valid prediction of the effectiveness of chemotherapeutic agents in individual head and neck squamous cell carcinoma (HNSCC) is desirable and might be achieved using ex vivo assays. METHODS: Three biopsies from each of 15 HNSCC were taken, minced and digested by collagenase. The digested HNSCC was added to serial dilutions of either cisplatin (CIS) or docetaxel (DTX), which were prepared under flavin-protecting conditions in ECM-coated microtiterplates. After 72-h incubation, cultures were methanol-fixed and Giemsa-stained. The cutoff concentration (COC; concentration completely suppressing colony formation) for epithelial cells (EC) and stromal cells (SC) was evaluated. RESULTS: 12/15 HNSCC (80%) were evaluable. Despite significant correlation of COC of CIS in respect of colony formation of EC or SC, no significant differences in response of individual HNSCC specimens were found in the t test for paired samples (p > 0.16). The same applied to DTX. However, EC and SC showed heterogeneity in chemoresponses leading to COC variability of more than one titration step in 44.1% (CIS) and 20% of HNSCC (DTX). No significant correlation between the COC of both cell populations was found in HNSCC specimens. CONCLUSIONS: The ex vivo chemoresponse of EC and SC of HNSCC must be analyzed separately.


Subject(s)
Cisplatin/pharmacology , Epithelial Cells/drug effects , Stromal Cells/drug effects , Taxoids/pharmacology , Adult , Aged , Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Docetaxel , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Epithelial Cells/pathology , Female , Head and Neck Neoplasms/pathology , Humans , Male , Middle Aged , Stromal Cells/pathology , Tumor Cells, Cultured , Tumor Stem Cell Assay
17.
Cell Cycle ; 6(7): 853-62, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17377499

ABSTRACT

Cks1 is a member of the cyclin-dependent kinase subunit family. These proteins are essential components of cyclin/cyclin-dependent kinase (cdk) complexes contributing to cell cycle control in all eukaryotes. Cks1 protein is found overexpressed in a number of tumors. Expression of Cks1 mRNA starts in late G1 reaching a peak in S/G2-phases of the cell cycle. We find that this expression pattern depends on transcriptional regulation and is controlled by a combination of a cell cycle-dependent element (CDE) together with a cell cycle genes homology region (CHR) in the Cks1 promoter. Furthermore, we observe Cks1 mRNA and protein to be downregulated after induced expression of the tumor suppressor p53. This repression is due to p53 downregulating transcription from the Cks1 promoter. p53-dependent repression is seen in a dose-dependent manner and in several cell types of different origin. In contrast to p53, its homologues p63 and p73 do not significantly repress transcription from the Cks1 promoter. The Cks1 promoter does not contain a p53 binding site. For some promoters the CCAAT box-binding transcription factor NF-Y had been implicated in p53-dependent repression. NF-Y is the main activator for Cks1 transcription but does not influence p53-dependent repression from the Cks1 promoter. Generally, the observation that the potential oncogene Cks1 is downregulated by the tumor suppressor p53 corresponds well with the idea that p53 employs multiple ways in order to halt the cell cycle.


Subject(s)
Carrier Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Transformation, Neoplastic/metabolism , Cyclin-Dependent Kinases/metabolism , Down-Regulation/genetics , Promoter Regions, Genetic/genetics , Regulatory Elements, Transcriptional/genetics , Tumor Suppressor Protein p53/metabolism , Animals , CCAAT-Binding Factor/genetics , CCAAT-Binding Factor/metabolism , CDC2-CDC28 Kinases , Carrier Proteins/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cyclin-Dependent Kinases/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic/genetics , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Tandem Repeat Sequences/genetics , Tumor Protein p73 , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
19.
Biochem Biophys Res Commun ; 320(3): 951-60, 2004 Jul 30.
Article in English | MEDLINE | ID: mdl-15240141

ABSTRACT

p55Cdc proteins participate in activation and timing of ubiquitin ligation by APC/C. Labeling of the substrates with ubiquitin leads to degradation of the cell cycle proteins through the proteasome in mitosis. Consistent with the phase in which the protein functions p55Cdc mRNA is expressed during the cell cycle starting in S phase with a maximum in G2/M. We analyzed the human p55Cdc promoter responsible for this expression pattern and found with SIRF (Cell-Cycle Site-Regulating p55Cdc/Fizzy-Transcription) a novel element which downregulates transcription in a cell cycle-dependent manner. Activation of gene transcription is independent of the SIRF element and NF-Y. The nucleotide sequence of SIRF is essentially identical in human, rat, and mouse p55Cdc whereas other parts of the promoter are not conserved. SIRF requires its natural promoter context for its regulatory function. With a length of 44 nucleotides this element is unusually long and may require a large protein complex for its regulation.


Subject(s)
Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Cycle/physiology , Genes, Regulator/physiology , Transcription, Genetic/physiology , Animals , Base Sequence , Cdc20 Proteins , Gene Expression Regulation/physiology , Mice , Molecular Sequence Data , NIH 3T3 Cells , Promoter Regions, Genetic/genetics , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL