Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-18465443

ABSTRACT

Mesenchymal stromal stem cells (MSC) can be found in almost any adult organ. They can be isolated and expanded within several weeks up to hundreds of millions of cells. The cell isolation based on the surface antigen expression may significantly enrich for the desired cell population and reduce the time required for cell expansion. MSC display a unique molecular signature which clearly discriminates them from other stem cell types. MSC can be differentiated into the cells of several lineages. Additionally, the unique biological properties of MSC are mediated by strong immunomodulatory activity and by paracrine mechanisms. Potential therapeutic applications of the cells require clinically compliant protocols for cell isolation and expansion. The therapeutic utility of MSC has been evaluated and found to be useful in several pre-clinical animal models as well as in clinical trials.


Subject(s)
Cell Culture Techniques , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Adult , Animals , Cell Differentiation/physiology , Clinical Trials as Topic , Disease Models, Animal , Humans
3.
Radiats Biol Radioecol ; 42(6): 604-7, 2002.
Article in English | MEDLINE | ID: mdl-12530134

ABSTRACT

The most important stage in the making of mutations is a reparation of different DNA damage, including the more deleterious double-strand DNA breaks (DSB). The first stage of adaptive response--fundamental antimutagenic cell reaction, purposeful to reparation for induced DSB repair--is investigated in present work. Non-radioactive in situ hybridization of biotin-labeled DNA probe was used to mark chromosome 1 pericentromeric regions (PR) in G0 human lymphocytes. It was shown that under 3-10 cGy (X-radiation, 160 kV) PR become displaced from a nucleus periphery to inner territory of a nucleus. The moving process realizes during several hours after an irradiation. As far as some non-specific gene repressors are co-localized with chromosome centromeric regions it is possible hypothesizes that the displacement cause changing expression of some genes. It is possible to propose that an absence of radiation induced chromosome locus displacement may be one of causes DSB repair disturbance. This hypothesis was tested by the model. It is assumed that one consequence of the underlying defect may be inappropriate involvement of cell's recombination machinery in the repair of DSB. We studied lymphocytes of patients with hereditary BRCA2 mutation. It is thought that this gene takes part in DSB repair. The significant differences of the PR moving between control samples and the cases were revealed under 10 cGy. Similar results were observed on lymphocytes of patients with Fanconi syndrome. Thus, abnormal moving of interphase nucleus chromosomes conditioned by low-dose irradiation may suggest on imperfect machinery of DSB repair, i.e. genetic risk. We realize that further investigations are needed for definitive conclusion.


Subject(s)
Chromosomes, Human, Pair 1/radiation effects , Interphase/radiation effects , DNA Damage , DNA Repair , Dose-Response Relationship, Radiation , Humans , In Situ Hybridization , Lymphocytes/radiation effects , Lymphocytes/ultrastructure
4.
Int J Cancer ; 93(6): 810-6, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11519042

ABSTRACT

Epstein-Barr virus (EBV) immortalized cells and Burkitt lymphoma cells have a completely different growth pattern and phenotype. EBV immortalized cells express a set of 11 viral genes to accommodate B cell activation and proliferation, whereas EBV-positive Burkitt lymphoma cells highly express the c-myc oncogene that is activated through translocation into 1 of the immunoglobulin loci and EBNA1 as the only viral protein. We have developed a primary human B cell line conditionally immortalized by Epstein-Barr virus in which the viral gene program responsible for the induction of proliferation can be switched on and off by the addition or withdrawal of estrogen (cell line EREB2-5). Starting from this cell line we have generated 2 daughter cell lines that proliferate in a c-myc dependent fashion, 1 with a highly active exogenous c-myc gene (cell line A1) and 1 with a regulatable c-myc gene that can be switched on by withdrawal and switched off by addition of tetracycline (cell line P493-6). The comparison of the 3 cell lines has allowed us to dissect the contribution of c-myc and EBV genes to the regulation of the growth pattern and expression of cell surface molecules. We show that MYC and EBNA2 (and their respective target genes) have opposing effects on the expression of several surface markers involved in B cell activation. We show that MYC contributes to the phenotype of Burkitt lymphoma cells by upregulating CD10 and CD38 and downregulating activation markers. The phenotype of the cells is determined on one hand by the absence of the viral gene products EBNA2 and LMP1 that mediate the phenotype of activated lymphoblasts and to a lesser extent by an active contribution of the c-myc gene.


Subject(s)
Antigens, CD , B-Lymphocytes/metabolism , Herpesvirus 4, Human/genetics , Proto-Oncogene Proteins c-myc/genetics , ADP-ribosyl Cyclase , ADP-ribosyl Cyclase 1 , Antigens, Differentiation/metabolism , Blotting, Western , Cell Division , Cell Line , Cell Separation , Dose-Response Relationship, Drug , Down-Regulation , Epstein-Barr Virus Nuclear Antigens/genetics , Flow Cytometry , Humans , Lymphocyte Activation , Membrane Glycoproteins , NAD+ Nucleosidase/metabolism , Neprilysin/metabolism , Phenotype , Recombinant Fusion Proteins/metabolism , Tetracycline/pharmacology , Time Factors , Transfection , Tumor Cells, Cultured , Up-Regulation , Viral Matrix Proteins/genetics , Viral Proteins
5.
Int J Cancer ; 92(2): 276-84, 2001 Apr 15.
Article in English | MEDLINE | ID: mdl-11291057

ABSTRACT

Cytological screening for cervical cancer or its precursors using Papanicolaou's smear test (Pap test) has been highly efficient to reduce the morbidity and mortality of cervical cancer. However, evaluation of the Pap test relies on subjective diagnostic parameters and is affected by a high rate of false-positive and false-negative results. More objective diagnostic parameters to identify truly dysplastic or neoplastic cells in cervical smears as well as in cervical biopsy samples would therefore avoid insecurity for many patients and the high screening costs associated with repeated testing. Cervical dysplasia is induced by persistent infections through high-risk types of human papillomaviruses (HPVs). Outgrowth of dysplastic lesions is triggered by increasing expression of two viral oncogenes, E6 and E7, which both interact with various cell cycle-regulating proteins. Among these is the retinoblastoma gene product pRB, which is inactivated by E7. pRB inhibits transcription of the cyclin-dependent kinase inhibitor gene p16(INK4a). Increasing expression of the viral oncogenes in dysplastic cervical cells might thus be reflected by increased expression of p16(INK4a). In line with this hypothesis, we observed marked overexpression of p16(INK4a) in all cervical intraepithelial neoplasm (CIN) I lesions (n = 47) except those associated with low-risk HPV types (n = 7), all CIN II lesions (n = 32), all CIN III lesions (n = 60) and 58 of 60 invasive cervical cancers. In contrast, no detectable expression of p16(INK4a) was observed in normal cervical epithelium (n = 42), inflammatory lesions (n = 48) and low-grade cervical lesions (CIN I) associated with low-risk HPV types (n = 7). Dysplastic cells could also be identified in cervical smears using a specific p16(INK4a) monoclonal antibody. These data demonstrate that p16(INK4a) is a specific biomarker to identify dysplastic cervical epithelia in sections of cervical biopsy samples or cervical smears.


Subject(s)
Biomarkers, Tumor/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Uterine Cervical Dysplasia/metabolism , Uterine Cervical Neoplasms/metabolism , Cyclin-Dependent Kinase Inhibitor p16/immunology , Female , Humans , Immunohistochemistry , Papillomaviridae/isolation & purification , Tumor Cells, Cultured , Uterine Cervical Dysplasia/pathology , Uterine Cervical Dysplasia/virology , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/virology
6.
Int J Cancer ; 87(6): 787-93, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-10956386

ABSTRACT

The product of the proto-oncogene c-myc (myc) is a potent activator of cell proliferation. In Burkitt lymphoma (BL), a human B-cell tumor, myc is consistently found to be transcriptionally activated by chromosomal translocation. The mechanisms by which myc promotes cell cycle progression in B-cells is not known. As a model for myc activation in BL cells, we have established a human EBV-EBNA1 positive B-cell line, P493-6, in which myc is expressed under the control of a tetracycline regulated promoter. If the expression of myc is switched off, P493-6 cells arrest in G0/G1 in the presence of serum. Re-expression of myc activates the cell cycle without inducing apoptosis. myc triggers the expression of cyclin D2, cyclin E and Cdk4, followed by the activation of cyclin E-associated kinase and hyper-phosphorylation of Rb. The transcription factor E2F-1 is expressed in proliferating and arrested cells at constant levels. The Cdk inhibitors p16, p21, p27 and p57 are expressed at low or not detectable levels in proliferating cells and are not induced after repression of myc. Ectopic expression of p16 inhibits cell cycle progression. These data suggest that myc triggers proliferation of P493-6 cells by promoting the expression of a set of cell cycle activators but not by inactivating cell cycle inhibitors.


Subject(s)
Burkitt Lymphoma/physiopathology , Cell Cycle Proteins/physiology , Cell Cycle/physiology , Neoplasm Proteins/physiology , Proto-Oncogene Proteins c-myc/physiology , Burkitt Lymphoma/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p16/physiology , Humans , Neoplasm Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Phosphorylation , Protein Synthesis Inhibitors/pharmacology , Proto-Oncogene Mas , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Retinoblastoma Protein/metabolism , Retinoblastoma-Like Protein p107 , Tetracycline/pharmacology , Tumor Cells, Cultured
7.
Mol Cell Biol ; 18(1): 409-19, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9418888

ABSTRACT

The minute virus of mice, an autonomous parvovirus, requires entry of host cells into the S phase of the cell cycle for its DNA to be amplified and its genes expressed. This work focuses on the P4 promoter of this parvovirus, which directs expression of the transcription unit encoding the parvoviral nonstructural polypeptides. These notably include protein NS1, necessary for the S-phase-dependent burst of parvoviral DNA amplification and gene expression. The activity of the P4 promoter is shown to be regulated in a cell cycle-dependent manner. At the G1/S-phase transition, the promoter is activated via a cis-acting DNA element which interacts with phase-specific complexes containing the cellular transcription factor E2F. It is inhibited, on the other hand, in cells arrested in G1 due to contact inhibition. This inhibitory effect is not observed in serum-starved cells. It is mediated in cis by cyclic AMP response elements (CREs). Unlike serum-starved cells, confluent cells accumulate the cyclin-dependent kinase inhibitor p27, suggesting that the switch from CRE-mediated activation to CRE-mediated repression involves the p27 protein. Accordingly, plasmid-driven overexpression of p27 causes down-modulation of promoter P4 in growing cells, depending on the presence of at least two functional CREs. No such effect is observed with two other cyclin-dependent kinase inhibitors, p16 and p21. Given the importance of P4-driven synthesis of protein NS1 in parvoviral DNA amplification and gene expression, the stringent S-phase dependency of promoter P4 is likely a major determinant of the absolute requirement of the minute virus of mice for host cell proliferation.


Subject(s)
Cell Cycle Proteins , Cyclic AMP Response Element-Binding Protein/genetics , Microtubule-Associated Proteins/genetics , Minute Virus of Mice/physiology , Promoter Regions, Genetic , Tumor Suppressor Proteins , Virus Integration , 3T3 Cells , Animals , Base Sequence , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/antagonists & inhibitors , Mice , Molecular Sequence Data , S Phase
8.
Cell Growth Differ ; 8(6): 699-710, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9186003

ABSTRACT

Treatment of mammalian cells by DNA-damaging agents leads to various cellular responses. At sufficiently high dosage, cisplatin blocks cell proliferation and finally kills cells; this effect is the basis for its widespread use as an anticancer drug. Cisplatin-treated cells arrest in the G1 phase of the cell cycle, most likely due to a signal generated by the stabilization of p53 and the subsequent induction of p21WAF-1/Cip1. We show here that cisplatin-treated mammalian cells accumulate normal levels of cyclin D1 and cyclin E but fail to produce cyclin A. The block to cyclin A gene expression occurs at the level of transcription and is mediated by an E2F binding site in the cyclin A promoter. It is shown here that, upon cisplatin treatment, transcriptionally active free E2F becomes limiting, coincident with the accumulation of hypophosphorylated species of the retinoblastoma protein family. Immunoprecipitation experiments suggest that the loss of free E2F results, at least in part, from the sequestration of E2F-4/DP-1 heterodimers by p107. A role for the kinase inhibitor p21WAF-1/Cip1 in repression of the cyclin A promoter is supported by our finding that ectopic expression of p21WAF-1/Cip1 is sufficient to inhibit transcription from the cyclin A gene, dependent on the E2F site. The data establish the E2F site in the human cyclin A promoter as a key target for the signaling pathway leading to G1 arrest in response to DNA damage by cisplatin and potentially other genotoxic agents.


Subject(s)
Antineoplastic Agents/pharmacology , Carrier Proteins , Cell Cycle Proteins , Cell Cycle/drug effects , Cisplatin/pharmacology , Cyclins/metabolism , DNA-Binding Proteins/metabolism , Down-Regulation , Transcription Factors/metabolism , 3T3 Cells/chemistry , 3T3 Cells/drug effects , Animals , Blotting, Northern , Blotting, Western , Cell Cycle/genetics , Cyclin D1 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/pharmacology , E2F Transcription Factors , E2F4 Transcription Factor , Enzyme Inhibitors/pharmacology , Fibroblasts/chemistry , Fibroblasts/drug effects , G1 Phase/drug effects , G1 Phase/genetics , Humans , Mice , Oncogene Proteins/metabolism , Phosphorylation , Plasmids , Promoter Regions, Genetic , RNA, Messenger , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1 , Transcription, Genetic , Transfection
9.
Oncogene ; 14(10): 1137-45, 1997 Mar 13.
Article in English | MEDLINE | ID: mdl-9121762

ABSTRACT

In this study we show, by immunofluorescence and electron microscopy immuno-gold labelling, that the major transforming protein of Human Papillomavirus type 16 E7 is associated with the nucleolus of cells derived from the HPV16-positive cervical carcinoma line CaSki. The E7 nucleolar staining appeared to be cell cycle dependent, being considerably reduced in the G2 phase. The total level of the protein in the cell, however, remained constant during all phases. We also show that the cellular protein Rb1, which is targeted by E7, is localised in the nucleus and nucleolus in CaSki cells. Thus, it is possible that the presence of E7 in the nucleolus correlates with a hypothetical function(s) of Rb1 in this particular intranuclear compartment. The nucleolar localisation of HPV16 E7 protein was also observed in the fission yeast Schizosaccharomyces pombe, suggesting that a targeting mechanism of HPV16 E7 protein into the nucleolus is common to both mammalian and yeast systems. Nucleolar localisation of HPV16 E7 protein may be independent from Rb1 since no Rb1 related proteins have been identified in fission yeast.


Subject(s)
Cell Nucleolus/metabolism , Oncogene Proteins, Viral/metabolism , Repressor Proteins , Schizosaccharomyces/metabolism , Uterine Cervical Neoplasms/metabolism , Antibodies, Monoclonal , Cell Cycle/physiology , Female , Humans , Immunohistochemistry , Microscopy, Electron , Microscopy, Fluorescence , Retinoblastoma Protein/analysis , Schizosaccharomyces/ultrastructure , Tumor Cells, Cultured , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/ultrastructure
10.
Verh Dtsch Ges Pathol ; 81: 233-9, 1997.
Article in English | MEDLINE | ID: mdl-9474875

ABSTRACT

Steroid hormones are frequently prescribed as anticontraceptive drugs to young women. Persistent papillomavirus infections particularly with high risk virus types are very common in younger women and were shown to be the strongest risk factor for the later development of cervical cancer. Steroid hormones interfere with persistent papillomavirus infections on various levels. They enhance the expression level of two viral genes, E6 and E7, which are required for the oncogenic activities of high risk papillomaviruses. In addition, they interfere with cellular gene functions involved in cell cycle regulation and programmed cell death, for example through inhibition of p53-mediated transcriptional transactivation of genes involved in cell cycle arrest and apoptosis. Furthermore, steroids inhibit the immunologically mediated resolution of minor HPV-induced cervical lesions, particularly through inhibition of major histocompatibility class I and class II antigen expression. These observations point to potent cocarcinogenic effects of steroid hormones in persistently papillomavirus infected individuals by enhancing the transforming activities of viral oncogenes and interfering with the efficient resolution of virus infected lesions. The clinical significance of these experimental observations requires careful analysis in prospective trials.


Subject(s)
Chromosome Aberrations , Estrogens/physiology , Oncogenes , Papillomaviridae/isolation & purification , Progesterone/physiology , Uterine Cervical Dysplasia/genetics , Uterine Cervical Neoplasms/genetics , Apoptosis , Cell Cycle , Contraceptives, Oral/adverse effects , Estrogens/adverse effects , Female , Genome, Viral , Humans , Oncogene Proteins, Viral/genetics , Papillomaviridae/genetics , Progesterone/adverse effects , Risk Factors , Uterine Cervical Dysplasia/etiology , Uterine Cervical Dysplasia/pathology , Uterine Cervical Dysplasia/virology , Uterine Cervical Neoplasms/etiology , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/virology
11.
Biochemistry (Mosc) ; 62(11): 1285-90, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9467852

ABSTRACT

Dynamics of telomere sequences are considered in normal and immortalized cells. Immortalized cells are suggested to be derived mainly from a special subpopulation of ontogenetic reserve cells. Their epigenetic program consists of autoregeneration during external stimulus for genome reorganization and corresponding appearance of genetic variants of cells. It is suggested that cells surviving the crisis stage contain a special signal sequence integrated into telomere DNA. Its elimination during shortening of DNA telomere sequences in dividing ontogenetic reserve cells is a signal for the cooperative transition of chromatin into a new steady state that corresponds to the epigenotype of immortalized cells. Localization of telomere DNA sequences in intrachromosomal "hot spots" reflects phylogenetic rearrangement of the genome.


Subject(s)
DNA/genetics , Telomere/genetics , Animals , Cell Division , Cell Line, Transformed , DNA Replication , Genetic Variation , Humans , Telomerase/metabolism
12.
Int J Cancer ; 68(4): 506-13, 1996 Nov 15.
Article in English | MEDLINE | ID: mdl-8945623

ABSTRACT

The E6 gene of tumor-associated types of human papillomaviruses codes for a functional antagonist of p53. Overexpression of E6 from heterologous promoters can block p53-mediated cellular responses to DNA damage, such as transcriptional stimulation of p53 target genes and cell-cycle arrest in G1. In contrast, genotoxic treatment of HPV-positive cancer cells, which express the E6 gene from chromosomally integrated viral copies, results in increased expression of the p53 target gene p21WAF1 and, in several cell lines, induction of G1 arrest. In the present study, we show that treatment with genotoxic agents, such as mitomycin C and cisplatin, leads to strong repression of viral E6/E7 oncogene expression in HPV16- and HPV18-positive cervical carcinoma cell lines. Kinetic analyses revealed that reduction of E6/E7 expression was not a prerequisite for induction of p21WAF1. We furthermore found that the apoptosis-promoting bax gene could be induced by genotoxic stress in some, but not all, HPV-positive cancer cell lines. Treatment with DNA-damaging agents eventually resulted in apoptotic cell death of HPV-positive cancer cells, irrespective of their capacity to induce the p53 target gene bax. These results support the notion that HPV-positive cancer cells can exhibit intact cellular responses to genotoxic stress, which may involve p53-dependent and -independent biochemical pathways. The ability of HPV-positive cancer cells to induce apoptotic cell death in response to DNA damage could provide a molecular explanation for the therapeutic effects of genotoxic agents in the treatment of cervical cancer.


Subject(s)
Apoptosis/drug effects , DNA Damage , DNA-Binding Proteins , Oncogene Proteins, Viral/genetics , Oncogenes , Repressor Proteins , Uterine Cervical Neoplasms/drug therapy , Cisplatin/pharmacology , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , Female , Humans , Mitomycin/pharmacology , Papillomavirus E7 Proteins , Tumor Cells, Cultured , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/virology
13.
Mol Cell Biol ; 16(10): 5302-12, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8816442

ABSTRACT

Terminally differentiated cells are characterized by permanent withdrawal from the cell cycle; they do not enter S phase even when stimulated by growth factors or retroviral oncogenes. We have shown, however, that the adenovirus E1A oncogene can reactivate the cell cycle in terminally differentiated cells. In this report, we describe the molecular events triggered by E1A in terminally differentiated skeletal muscle cells. We found that in myotubes infected with the adenovirus mutant dl520, 12S E1A bypasses the early G1 phase and activates the expression of late-G1 genes, such as the cyclin E and cyclin A genes, cdk2, PCNA, and B-myb. Of these, the cyclin E gene and cdk2 were significantly overexpressed in comparison with levels in proliferating, undifferentiated myoblasts. p130 and pRb were phosphorylated before the infected myotubes entered S phase, despite the high expression of the cyclin-dependent kinase inhibitor p21, and E2F was released. Our results suggest that one of the mechanisms that E1A uses to overcome the proliferative block of terminally differentiated cells involves coordinated overexpression of cyclin E and cdk2. Following E1A expression, the myogenic transcription factors MyoD and myogenin and the muscle-specific structural genes encoding muscle creatine kinase and myosin heavy chain were downregulated. The muscle regulatory factors were also silenced in myotubes infected with adenovirus E1A mutants incapable of reactivating the cell cycle in terminally differentiated muscle cells. Thus, the suppression of the differentiation program is not a consequence of cell cycle reactivation in myotubes, and it is induced by an independent mechanism. Our results show that E1A reactivates the cell cycle and suppresses tissue-specific gene expression in terminally differentiated muscle cells, thus causing dedifferentiation.


Subject(s)
Adenovirus E1A Proteins/physiology , Carrier Proteins , Cell Cycle Proteins , Cell Cycle , Cell Differentiation , DNA-Binding Proteins , Gene Expression Regulation , Gene Expression , Muscle, Skeletal/cytology , Adenovirus E1A Proteins/biosynthesis , Animals , Cell Cycle/genetics , Cell Differentiation/genetics , Cell Division , Cell Line , Cyclins/biosynthesis , E2F Transcription Factors , G1 Phase , Genetic Markers , Kinetics , Mice , Recombinant Proteins/biosynthesis , Retinoblastoma Protein/biosynthesis , Retinoblastoma-Binding Protein 1 , Retroviridae , Transcription Factor DP1 , Transcription Factors/metabolism , Transfection
14.
Oncogene ; 13(5): 1027-35, 1996 Sep 05.
Article in English | MEDLINE | ID: mdl-8806692

ABSTRACT

Growth of cervical carcinoma cells depends on continuous expression of high risk type human papillomavirus oncogenes E6 and E7. E6 destabilizes p53, a tumor-suppressive transcription factor, which activates expression of the inhibitor of cell cycle progression p21 and other genes. E6-mediated p53 degradation can therefore result in cell cycle deregulation. It has, however, not yet been determined whether p53 inactivation is sufficient to provoke cell cycle progression in established cervical carcinoma cells. Moreover, it has not yet been clarified whether E6 confers additional p53-independent growth stimuli in cancer cells. To address these questions, we analysed p53 functions in SW 756 cervical cancer cells in which the expression of endogenous HPV 18 E6-E7 genes can be downregulated by dexamethasone. This results in significantly increased p53 levels and subsequent cell cycle arrest in the Gz phase. Surprisingly, p53 activities were suppressed rather than enhanced in these cervical cancer cells. However, if high risk papillomavirus type 16 E6 genes, including a mutant which does not degrade p53, were expressed in dexamethasone-treated SW 756 cells with suppressed endogenous HPV type 18 E6-E7 expression, the cells reentered the cell cycle even in the absence of a cooperating viral E7 gene. In contrast, the non oncogenic papillomavirus type 6 E6 gene did not release the cells from growth arrest under these conditions. These data indicate that suppression of p53 functions is not sufficient to provoke cell cycle progression in E6-E7-depleted cervical cancer cells and point to a p53-independent mitotic activity to oncogenic papillomavirus type E6 genes in cervical carcinoma cells.


Subject(s)
DNA-Binding Proteins , Oncogene Proteins, Viral/genetics , Repressor Proteins , Tumor Suppressor Protein p53/genetics , Uterine Cervical Neoplasms/genetics , Amino Acid Sequence , Carcinoma/genetics , Carcinoma/pathology , Carcinoma/virology , Cell Cycle/genetics , Cell Division/drug effects , Cell Division/genetics , Clone Cells , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , Dexamethasone/pharmacology , Female , G1 Phase/genetics , Gene Expression Regulation, Neoplastic , Gene Expression Regulation, Viral , Genes, Reporter , Humans , Molecular Sequence Data , Mutation , Oncogene Proteins, Viral/biosynthesis , Plasmids/genetics , Trans-Activators , Transcription, Genetic , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/virology
15.
Mol Cell Biol ; 16(7): 3401-9, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8668155

ABSTRACT

Cyclin E controls progression through the G1 phase of the cell cycle in mammalian fibroblasts and potentially in many other cell types. Cyclin E is a rate-limiting activator of cdk2 kinase in late G1. The abundance of cyclin E is controlled by phase-specific fluctuations in the mRNA level; in mammalian fibroblasts, mRNA is not detected under conditions of serum starvation and is accumulated upon serum stimulation, with expression starting in mid-G1. Here, we report the cloning of the murine cyclin E promoter. We isolated a 3.8-kb genomic fragment that contains several transcriptional start sites and confers cell cycle regulation on a luciferase reporter gene. This fragment also supports transcriptional activation by adenovirus E1A, a known upstream regulator of cyclin E gene expression. An E2F binding site which is required for G1-specific activation of the cyclin E promoter in synchronized NIH 3T3 cells was identified in this fragment.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , Cyclins/biosynthesis , Cyclins/genetics , DNA-Binding Proteins , Gene Expression Regulation , Promoter Regions, Genetic , Transcription Factors/metabolism , 3T3 Cells , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Cloning, Molecular , E2F Transcription Factors , Humans , Mammals , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Oligodeoxyribonucleotides , Point Mutation , Recombinant Proteins/biosynthesis , Retinoblastoma-Binding Protein 1 , Sequence Homology, Nucleic Acid , Transcription Factor DP1 , Transcription, Genetic
16.
Oncogene ; 12(12): 2549-54, 1996 Jun 20.
Article in English | MEDLINE | ID: mdl-8700513

ABSTRACT

Adenovirus E1A proteins can induce quiescent cells to enter S-phase and also affect the expression of cellular genes including various cell cycle regulators. Here we show that human cdc25A, a tyrosine phosphatase involved in regulation of the G1/S-phase transition of the cell cycle, is a target of the adenovirus E1A protein in virus-infected human fibroblasts. Expression of E1A in quiescent fibroblasts leads to a rapid increase in cdc25A phosphatase activity and also increases both cdc25A and cyclin E gene expression. Inhibition of cdc25A function by antibody injection prevents virus-induced entry into S-phase. These results indicate that induction of high levels of cdc25A and its potential positive regulator cyclin E mediates the ability of E1A to induce S-phase in the presence of antiproliferative signals.


Subject(s)
Adenovirus E1A Proteins/genetics , Cell Cycle Proteins/metabolism , Phosphoprotein Phosphatases/metabolism , S Phase/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/immunology , Cell Division/genetics , Cyclins/genetics , Fibroblasts/enzymology , Fibroblasts/virology , Gene Expression Regulation, Viral , Humans , Mutation , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/immunology , RNA, Messenger/biosynthesis , cdc25 Phosphatases
17.
J Virol ; 70(4): 2637-42, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8642699

ABSTRACT

Using the infection of quiescent human fibroblasts with adenovirus type 5 and various deletion mutants, we show that E1A can stimulate transcription of the cyclin A gene in the absence of exogenous growth factors. Required for this activity is conserved region 2 (CR2), while both the N-terminal part of E1A and CR1 are dispensable. This indicates that activation of cyclin A gene expression requires the binding of E1A to p107, while binding to either pRB or p300 is not involved in transcriptional activation. We demonstrate that p107 represses the cyclin A promoter through its cell cycle-regulatory E2F binding site and that 12S E1A can activate the cyclin A promoter, essentially by counteracting its repression by p107. Since Cr2 is required for cell transformation, transcriptional activation of the cyclin A gene by E1A appears to be important for its capacity to override control of cellular growth.


Subject(s)
Adenoviruses, Human/metabolism , Cyclins/genetics , Gene Expression Regulation, Viral , Nuclear Proteins/metabolism , 3T3 Cells , Animals , Binding Sites , Cell Line , Humans , Mice , Promoter Regions, Genetic , RNA, Messenger/metabolism , Retinoblastoma-Like Protein p107 , Transcriptional Activation
18.
Proc Natl Acad Sci U S A ; 92(24): 11264-8, 1995 Nov 21.
Article in English | MEDLINE | ID: mdl-7479977

ABSTRACT

Cyclin A is involved in the control of S phase and mitosis in mammalian cells. Expression of the cyclin A gene in nontransformed cells is characterized by repression of its promoter during the G1 phase of the cell cycle and its induction at S-phase entry. We show that this mode of regulation is mediated by the transcription factor E2F, which binds to a specific site in the cyclin A promoter. It differs from the prototype E2F site in nucleotide sequence and protein binding; it is bound by E2F complexes containing cyclin E and p107 but not pRB. Ectopic expression of cyclin D1 triggers premature activation of the cyclin A promoter by E2F, and this effect is blocked by the tumor suppressor protein p16INK4.


Subject(s)
Cell Cycle Proteins , Cell Cycle , Cyclins/genetics , Cyclins/physiology , Oncogene Proteins/physiology , Promoter Regions, Genetic , Proto-Oncogene Proteins , Transcription Factors/metabolism , 3T3 Cells , Animals , Base Sequence , Carrier Proteins/physiology , Cyclin D1 , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase Inhibitor p16 , Cyclin-Dependent Kinases/physiology , DNA-Binding Proteins/metabolism , E2F Transcription Factors , Gene Expression , Mice , Molecular Sequence Data , Oligodeoxyribonucleotides/chemistry , RNA, Messenger/genetics , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1 , Transcription, Genetic , Transcriptional Activation
19.
J Virol ; 69(10): 6389-99, 1995 Oct.
Article in English | MEDLINE | ID: mdl-7666540

ABSTRACT

To investigate E7-dependent biochemical changes which are involved in cellular transformation, we analyzed the influence of human papillomavirus type 16 (HPV-16) E7 on the expression of cell cycle regulatory proteins. Expression of E7 in established rodent fibroblasts (NIH 3T3), which was shown to be sufficient for transformation of these cells, leads to constitutive expression of the cyclin E and cyclin A genes in the absence of external growth factors. Surprisingly, expression of the cyclin D1 gene, which encodes a major regulator of G1 progression, is unaltered in E7-transformed cells. In transient transfection experiments, the cyclin A gene promoter is activated by E7 via an E2F binding site. In 14/2 cells, which were used as a model system to analyze the role of HPV-16 E7 in the transformation of primary cells, we observed rapid E7-dependent activation of cyclin E gene expression, which can be uncoupled from activation of the cyclin A gene, since the latter requires additional protein synthesis. E7-driven induction of cyclin E and cyclin A gene expression was accompanied by an increase in the associated kinase activities. Two domains of the E7 oncoprotein, which are designated cd1 and cd2, are essential for transformation of rodent fibroblasts. It is shown here that growth factor-independent expression of the cyclin E gene requires cd2 but not cd1, while activation of cyclin A gene expression requires cd1 function in addition to that of cd2. These data suggest that cyclin A gene expression is controlled by two distinct negative signals, one of which also restricts expression of the cyclin E gene. The ability of E7 to separately override each of these inhibitory signals, via cd1 and cd2, cosegregates with its ability to fully transform rodent fibroblasts. Unlike serum growth factors, E7 induces S-phase entry without activating cyclin D1 gene expression, in keeping with the finding that cyclin D1 function is not required in cells transformed by DNA tumor viruses.


Subject(s)
Cell Transformation, Neoplastic , Cyclin-Dependent Kinases/metabolism , Cyclins/biosynthesis , Gene Expression Regulation, Viral , Oncogene Proteins, Viral/metabolism , Papillomaviridae/metabolism , 3T3 Cells , Animals , Blotting, Western , Cell Cycle/drug effects , Cell Division/drug effects , Cell Line , Dexamethasone/pharmacology , Kinetics , Mice , Oncogene Proteins, Viral/biosynthesis , Papillomaviridae/genetics , Papillomavirus E7 Proteins , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Time Factors , Transfection
20.
Oncogene ; 10(12): 2421-5, 1995 Jun 15.
Article in English | MEDLINE | ID: mdl-7784093

ABSTRACT

Expression of the cyclin D1 gene is induced when quiescent fibroblasts are stimulated to reenter the cell cycle by addition of growth factors. Moderate ectopic expression of cyclin D1 in early G1 facilitates progression through G1. When transiently overexpressed at the G1/S boundary, cyclin D1 prevents S phase entry, suggesting a dual role for this protein in cellular growth control. It was shown that the retinoblastoma protein (pRB) can activate cyclin D1 gene expression; furthermore, there is evidence that expression of the cyclin D1 gene is down-regulated by the SV40 large T and adenovirus E1A genes, both of which were shown to target pRB. We now report that in diploid human fibroblasts functional inactivation of pRB by adenovirus E1A is not sufficient for efficient repression of cyclin D1 gene expression, since the E1B gene product, in addition to E1A, is required for repression of the cyclin D1 gene. Since E1B was shown to target p53, we investigated the role of p53 for expression of the cyclin D1 gene. In a cell line with temperature-sensitive p53, cyclin D1 is moderately expressed at the restrictive temperature. Induction of p53 function by temperature shift leads to an increase of cyclin D1 mRNA and protein, parallel to the activation of p21WAF-1/CIP1 gene expression in this system. When the capability of adenovirus gene products to affect expression of either gene was analysed, we found that infection of Ad5 drastically reduced cyclin D1 and p21WAF-1/CIP1 gene expression in cells where p53 function is limiting. Under these conditions E1A and E1B cooperate to reduce the cyclin D1 level, while p21WAF-1/CIP1 expression was found insensitive to E1A expression. In cells containing elevated p53 function, modulation of gene expression by E1B was severely compromised; under these conditions, expression of E1A reduced expression of cyclin D1 without affecting p21WAF-1/CIP1. The data suggest that E1A and E1B cooperate to inhibit expression of cyclin D1 and identify the cyclin D1 gene as a new downstream target for p53.


Subject(s)
Adenovirus E1A Proteins/physiology , Adenoviruses, Human/genetics , Cyclins/metabolism , Fibroblasts/metabolism , Genes, p53/physiology , Oncogene Proteins/metabolism , Oncogenes/physiology , Retinoblastoma Protein/metabolism , Adenovirus E1A Proteins/metabolism , Adenovirus E1B Proteins/metabolism , Adenovirus E1B Proteins/physiology , Cyclin D1 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , Gene Expression Regulation, Viral , Humans , Oncogene Proteins/genetics , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL
...