Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Microbiol ; 8(10): 1591-600, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16984414

ABSTRACT

Staphylococcal alpha-toxin is an archetypal killer protein that homo-oligomerizes in target cells to create small transmembrane pores. The membrane-perforating beta-barrel motif is a conserved attack element of cytolysins of Gram-positive and Gram-negative bacteria. Following the recognition that nucleated cells can survive membrane permeabilization, a profile of abundant transcripts was obtained in transiently perforated keratinocytes. Several immediate early genes were found to be upregulated, reminiscent of the cellular response to growth factors. Cell cycle analyses revealed doubling of S + G2/M phase cells 26 h post toxin treatment. Determination of cell counts uncovered that after an initial drop, numbers increased to exceed the controls after 2 days. A non-lytic alpha-toxin mutant remained without effect. The alpha-toxin pore is too small to allow egress of cytosolic growth factors, and evidence was instead obtained for growth signalling via the epidermal growth factor receptor (EGFR). Inhibition of the EGFR or of EGFR-proligand-processing blocked the mitogenic effect of alpha-toxin. Western blots with phospho-specific antibodies revealed activation of the EGFR, and of the adapter protein Shc. Immediate early response and proliferation upon transient plasma membrane pore formation by bacterial toxins may represent a novel facet of the complex interaction between pathogen and host.


Subject(s)
Bacterial Toxins , ErbB Receptors/metabolism , Hemolysin Proteins , Keratinocytes/metabolism , Staphylococcus aureus/physiology , Adaptor Proteins, Signal Transducing/metabolism , Blotting, Western , Cell Cycle , Cell Line , Cell Line, Transformed , Cell Proliferation , Cytotoxins/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression Profiling , Humans , Keratinocytes/cytology , Keratinocytes/microbiology , Mitogens/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Shc Signaling Adaptor Proteins , Signal Transduction , Src Homology 2 Domain-Containing, Transforming Protein 1 , Transfection
2.
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
3.
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
4.
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
5.
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
6.
J Biol Chem ; 278(9): 6642-50, 2003 Feb 28.
Article in English | MEDLINE | ID: mdl-12482752

ABSTRACT

The CCAAT box is one of the most common elements in eukaryotic promoters and is activated by NF-Y, a conserved trimeric transcription factor with histone-like subunits. Usually one CCAAT element is present in promoters at positions between -60 and -100, but an emerging class of promoters harbor multiple NF-Y sites. In the triple CCAAT-containing cyclin B2 cell-cycle promoter, all CCAAT boxes, independently from their NF-Y affinities, are important for function. We investigated the relationships between NF-Y and p300. Chromatin immunoprecipitation analysis found that NF-Y and p300 are bound to the cyclin B2 promoter in vivo and that their binding is regulated during the cell cycle, positively correlating with promoter function. Cotransfection experiments determined that the coactivator acts on all CCAAT boxes and requires a precise spacing between the three elements. We established the order of in vitro binding of the three NF-Y complexes and find decreasing affinities from the most distal Y1 to the proximal Y3 site. Binding of two or three NF-Y trimers with or without p300 is not cooperative, but association with the Y1 and Y2 sites is extremely stable. p300 favors the binding of NF-Y to the weak Y3 proximal site, provided that a correct distance between the three CCAAT is respected. Our data indicate that the precise spacing of multiple CCAAT boxes is crucial for coactivator function. Transient association to a weak site might be a point of regulation during the cell cycle and a general theme of multiple CCAAT box promoters.


Subject(s)
CCAAT-Binding Factor/metabolism , Cyclin B/genetics , Nuclear Proteins/metabolism , Trans-Activators/metabolism , 3T3 Cells , Animals , Base Sequence , Cell Cycle , Cell Line , Chromatin/metabolism , Cyclin B/chemistry , Cyclin B2 , Dimerization , E1A-Associated p300 Protein , Escherichia coli/metabolism , Humans , Insecta , Luciferases/metabolism , Mice , Models, Biological , Molecular Sequence Data , Mutation , Phosphorylation , Plasmids/metabolism , Precipitin Tests , Promoter Regions, Genetic , Protein Binding , Transcriptional Activation , Transfection , Tumor Cells, Cultured
7.
Nucleic Acids Res ; 30(9): 1967-76, 2002 May 01.
Article in English | MEDLINE | ID: mdl-11972334

ABSTRACT

The cdc25C phosphatase participates in regulating transition from the G2 phase of the cell cycle to mitosis by dephosphorylating cyclin-dependent kinase 1. The tumor suppressor p53 down-regulates expression of cdc25C as part of G2/M checkpoint control. Transcription of cdc25C oscillates during the cell cycle with no expression in resting cells and maximum transcription in G2. We had identified earlier a new mechanism of cell cycle-dependent transcription that is regulated by a cell cycle-dependent element (CDE) in conjunction with a cell cycle genes homology region (CHR). The human cdc25C gene was the first example. CDE/CHR tandem elements have since been found in promoters of many cell cycle genes. Here we show that the mouse cdc25C gene is regulated by a CHR but does not hold a CDE. Therefore, it is the first identified gene with CHR-dependent transcriptional regulation during the cell cycle not relying on a CDE located upstream of it. The CHR leads to repression of cdc25C transcription early in the cell cycle and directs a release of this repression in G2. Furthermore, we find that this CHR can cooperate in cell cycle-dependent transcription with elements placed directly upstream of it binding E2F, Sp1 or Sp3 transcription factors.


Subject(s)
Cell Cycle Proteins/genetics , DNA-Binding Proteins/metabolism , Response Elements , Sp1 Transcription Factor/metabolism , Transcription Factors/metabolism , cdc25 Phosphatases/genetics , 3T3 Cells , Amino Acid Sequence , Animals , Binding Sites , CCAAT-Binding Factor/metabolism , Cell Cycle , Consensus Sequence , Down-Regulation , E2F Transcription Factors , Humans , Mice , Molecular Sequence Data , Promoter Regions, Genetic , Sequence Homology, Amino Acid , Sp3 Transcription Factor , Transcriptional Activation , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...