Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 113
Filter
1.
Nat Genet ; 25(1): 47-54, 2000 May.
Article in English | MEDLINE | ID: mdl-10802655

ABSTRACT

The p73 protein, a homologue of the tumour-suppressor protein p53, can activate p53-responsive promoters and induce apoptosis in p53-deficient cells. Here we report that some tumour-derived p53 mutants can bind to and inactivate p73. The binding of such mutants is influenced by whether TP53 (encoding p53) codon 72, by virtue of a common polymorphism in the human population, encodes Arg or Pro. The ability of mutant p53 to bind p73, neutralize p73-induced apoptosis and transform cells in cooperation with EJ-Ras was enhanced when codon 72 encoded Arg. We found that the Arg-containing allele was preferentially mutated and retained in squamous cell tumours arising in Arg/Pro germline heterozygotes. Thus, inactivation of p53 family members may contribute to the biological properties of a subset of p53 mutants, and a polymorphic residue within p53 affects mutant behaviour.


Subject(s)
Mutagenesis, Site-Directed , Polymorphism, Genetic , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Alleles , Arginine/genetics , Carcinoma, Squamous Cell/genetics , Cell Line , Codon/genetics , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Genes, Tumor Suppressor , Genes, p53 , Genetic Carrier Screening , Germ-Line Mutation , Humans , Macromolecular Substances , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Nuclear Proteins/physiology , Proline/genetics , Protein Binding/genetics , Protein Conformation , Tumor Cells, Cultured , Tumor Protein p73 , Tumor Suppressor Protein p53/physiology , Tumor Suppressor Proteins
2.
Curr Opin Cell Biol ; 13(3): 332-7, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11343904

ABSTRACT

Loss of the p53 tumor suppressor pathway contributes to the development of most human cancers. p53 is a nuclear protein that functions as a regulator of transcription. Significant advances have been made recently in our understanding of how p53 function is regulated and the mechanisms by which p53 mediates its effects.


Subject(s)
Apoptosis/physiology , Cell Compartmentation/physiology , Neoplasms/pathology , Neoplasms/physiopathology , Transcription, Genetic/physiology , Tumor Suppressor Protein p53/metabolism , Active Transport, Cell Nucleus/physiology , Animals , Cell Cycle/physiology , Cell Division/physiology , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/physiology , Humans , Mutation/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic/genetics , Tumor Suppressor Protein p53/genetics
3.
Trends Cell Biol ; 10(5): 197-202, 2000 May.
Article in English | MEDLINE | ID: mdl-10754563

ABSTRACT

The tumour-suppressor protein p53 has recently been shown to belong to a family that includes two structurally related proteins, p63 and p73. Although all three proteins share similar transcriptional functions and the ability to induce apoptosis, each of them appears to play a distinct role in development and tumour suppression. In order for cell division to occur, the antiproliferative activities of these proteins must be tightly controlled, and exciting advances have been made in our understanding of the pathways involved in regulating p53 activity.


Subject(s)
Membrane Proteins , Trans-Activators , Tumor Suppressor Protein p53/physiology , Animals , Apoptosis , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Genes, p53 , Humans , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Phosphoproteins/genetics , Phosphoproteins/physiology , Protein Binding , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2 , Transcription Factors , Tumor Protein p73 , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins
4.
Oncogene ; 26(9): 1306-16, 2007 Feb 26.
Article in English | MEDLINE | ID: mdl-17322916

ABSTRACT

Over the years, p53 has been shown to sit at the centre of an increasingly complex web of incoming stress signals and outgoing effector pathways. The number and diversity of stress signals that lead to p53 activation illustrates the breadth of p53's remit - responding to a wide variety of potentially oncogenic insults to prevent tumour development. Interestingly, different stress signals can use different and independent pathways to activate p53, and there is some evidence that different stress signals can mediate different responses. How each of the responses to p53 contributes to inhibition of malignant progression is beginning to be clarified, with the hope that identification of responses that are key to tumour suppression will allow a more focused and effective search for new therapeutic targets. In this review, we will highlight some recently identified roles for p53 in tumour suppression, and discuss some of the numerous mechanisms through which p53 can be regulated and activated.


Subject(s)
Oxidative Stress , Tumor Suppressor Protein p53/physiology , Humans , Neoplasms/physiopathology , Neoplasms/therapy , Signal Transduction
5.
Curr Opin Genet Dev ; 6(1): 12-8, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8791489

ABSTRACT

The cell cycle arrest and apoptotic functions of p53 both contribute to the role of this tumour suppressor protein in preventing replication of cells suffering DNA damage. Although the ability of p53 to function as a sequence-specific transcription factor appears to be directly and causally linked to the implementation of an arrest at the G1 stage of the cell cycle, the contribution of transcriptional activation to the apoptotic response is less clear. It seems likely that several p53 activities, both transcriptionally dependent and transcriptionally independent, can play a role in mediating cell death. The requirement for each of these functions appears to depend on the cell type, the cell environment and other genetic alterations already sustained by the cell in which p53 function is activated.


Subject(s)
Apoptosis/physiology , Genes, p53/physiology , Signal Transduction/physiology , Animals , Cell Death/physiology , G1 Phase/physiology , Humans , Transcription, Genetic
6.
Curr Biol ; 10(9): 539-42, 2000 May 04.
Article in English | MEDLINE | ID: mdl-10801444

ABSTRACT

The MDM2 protein targets the p53 tumor suppressor for ubiquitin-dependent degradation [1], and can function both as an E3 ubiquitin ligase [2] and as a regulator of the subcellular localization of p53 [3]. Oncogene activation stabilizes p53 through expression of the ARF protein (p14(ARF) in humans, p19(ARF) in the mouse) [4], and loss of ARF allows tumor development without loss of wild-type p53 [5] [6]. ARF binds directly to MDM2, and prevents MDM2 from targeting p53 for degradation [6] [7] [8] [9] by inhibiting the E3 ligase activity of MDM2 [2] and preventing nuclear export of MDM2 and p53 [10] [11]. Interaction between ARF and MDM2 results in the localization of both proteins to the nucleolus [12] [13] [14] through nucleolar localization signals (NoLS) in ARF and MDM2 [11] [12] [13] [14]. Here, we report a new NoLS within the highly conserved amino-terminal 22 amino acids of p14(ARF), a region that we found could interact with MDM2, relocalize MDM2 to the nucleolus and inhibit the ability of MDM2 to degrade p53. In contrast, the carboxy-terminal fragment of p14(ARF), which contains the previously described NoLS [11], did not drive nucleolar localization of MDM2, although this region could bind MDM2 and weakly inhibit its ability to degrade p53. Our results support the importance of nucleolar sequestration for the efficient inactivation of MDM2. The inhibition of MDM2 by a small peptide from the amino terminus of p14(ARF) might be exploited to restore p53 function in tumors.


Subject(s)
Neoplasm Proteins/metabolism , Nuclear Localization Signals , Nuclear Proteins/metabolism , Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Protein p53 , Binding Sites , Humans , Proto-Oncogene Proteins c-mdm2 , Recombinant Fusion Proteins/metabolism , Tumor Suppressor Protein p14ARF
7.
Mol Cell Biol ; 17(1): 460-8, 1997 Jan.
Article in English | MEDLINE | ID: mdl-8972227

ABSTRACT

The p53 tumor suppressor protein is activated in cells in response to DNA damage and prevents the replication of cells sustaining genetic damage by inducing a cell cycle arrest or apoptosis. Activation of p53 is accompanied by stabilization of the protein, resulting in accumulation to high levels within the cell. p53 is normally degraded through the proteasome following ubiquitination, although the mechanisms which regulate this proteolysis in normal cells and how the p53 protein becomes stabilized following DNA damage are not well understood. We show here that p53 can also be a substrate for cleavage by the calcium-activated neutral protease, calpain, and that a preferential site for calpain cleavage exists within the N terminus of the p53 protein. Treatment of cells expressing wild-type p53 with an inhibitor of calpain resulted in the stabilization of the p53 protein. By contrast, in vitro or in vivo degradation mediated by human papillomavirus E6 protein was unaffected by the calpain inhibitor, indicating that the stabilization did not result from inhibition of the proteasome. These results suggest that calpain cleavage plays a role in regulating p53 stability.


Subject(s)
Calpain/metabolism , Repressor Proteins , Tumor Suppressor Protein p53/metabolism , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Adenylyl Imidodiphosphate/pharmacology , Amino Acid Sequence , Breast Neoplasms , Calpain/antagonists & inhibitors , Carcinoma , Chelating Agents/pharmacology , Cysteine Proteinase Inhibitors/pharmacology , Edetic Acid/pharmacology , Humans , Molecular Sequence Data , Mutation , Oncogene Proteins, Viral/metabolism , Papillomaviridae , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
8.
Mol Cell Biol ; 18(7): 3692-8, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9632751

ABSTRACT

Suppression of tumor cell growth by p53 results from the activation of both apoptosis and cell cycle arrest, functions which have been shown to be separable activities of p53. We have characterized a series of p53 mutants with amino acid substitutions at residue 175 and show that these mutants fall into one of three classes: class I, which is essentially wild type for apoptotic and cell cycle arrest functions; class II, which retains cell cycle arrest activity but is impaired in the induction of apoptosis; and class III, which is defective in both activities. Several residue 175 mutants which retain cell cycle arrest function have been detected in cancers, and we show that these have lost apoptotic function. Furthermore, several class II mutants have been found to be temperature sensitive for apoptotic activity while showing constitutive cell cycle arrest function. Taken together, these mutants comprise an excellent system with which to investigate the biochemical nature of p53-mediated apoptosis, the function of principal importance in tumor suppression. All of the mutants that showed loss of apoptotic function also showed defects in the activation of promoters from the potential apoptotic targets Bax and the insulin-like growth factor-binding protein 3 gene (IGF-BP3), and a correlation between full apoptotic activity and activation of both of these promoters was also seen with the temperature-sensitive mutants. However, a role for additional apoptotic activities of p53 was suggested by the observation that some mutants retained significant apoptotic function despite being impaired in the activation of Bax- and IGF-BP3-derived promoters. In contrast to the case of transcriptional activation, a perfect correlation between transcriptional repression of the c-fos promoter and the ability to induce apoptosis was seen, although the observation that Bax expression induced a similar repression of transcription from this promoter suggests that this may be a consequence, rather than a cause, of apoptotic death.


Subject(s)
Apoptosis , Cell Cycle , Mutagenesis, Site-Directed , Tumor Suppressor Protein p53/genetics , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , Humans , Insulin-Like Growth Factor Binding Protein 3/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-fos/genetics , Temperature , Transcriptional Activation , Tumor Cells, Cultured , Tumor Suppressor Protein p53/physiology , bcl-2-Associated X Protein
9.
Mol Cell Biol ; 20(9): 3224-33, 2000 May.
Article in English | MEDLINE | ID: mdl-10757806

ABSTRACT

The p53 tumor suppressor is activated by many diverse stress signals through mechanisms that result in stabilization and accumulation of the p53 protein. p53 is normally degraded through the proteasome following interaction with MDM2, which both functions as a ubiquitin ligase for p53 and shuttles to the cytoplasm, where p53 degradation occurs. Stabilization of p53 in response to stress is associated with inhibition of MDM2-mediated degradation, which has been associated with phosphorylation of p53 in response to DNA damage or activation of ARF. In this study we show distinct responses, as measured by phosphorylation, transcriptional activity, and subcellular localization, of p53 stabilized by different activating signals. Although normal cells and wild-type p53-expressing tumor cells showed similar responses to actinomycin D and camptothecin treatment, the transcriptional activity of stabilized p53 induced by deferoxamine mesylate, which mimics hypoxia, in normal cells was lost in all three tumor cell lines tested. Our results show that multiple pathways exist to stabilize p53 in response to different forms of stress, and they may involve down-regulation of MDM2 expression or regulation of the subcellular localization of p53 or MDM2. Loss of any one of these pathways may predispose cells to malignant transformation, although reactivation of p53 might be achieved through alternative pathways that remain functional in these tumor cells.


Subject(s)
Nuclear Proteins , Stress, Physiological , Tumor Suppressor Protein p53/metabolism , Blotting, Northern , Camptothecin/pharmacology , Cells, Cultured , Chelating Agents/pharmacology , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , Dactinomycin/pharmacology , Deferoxamine/pharmacology , Down-Regulation , Enzyme Inhibitors/pharmacology , Fibroblasts/metabolism , Humans , Immunoblotting , Microscopy, Fluorescence , Nucleic Acid Synthesis Inhibitors/pharmacology , Phosphorylation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2 , Serine/metabolism , Time Factors , Transcription, Genetic , Tumor Cells, Cultured
10.
Mol Cell Biol ; 16(9): 4952-60, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8756654

ABSTRACT

The p53 tumor suppressor protein is a sequence-specific transcriptional activator, a function which contributes to cell cycle arrest and apoptosis induced by p53 in appropriate cell types. Analysis of a series of p53 point mutants has revealed the potential for selective loss of the ability to transactivate some, but not all, cellular p53-responsive promoters. p53 175P and p53 181L are tumor-derived p53 point mutants which were previously characterized as transcriptionally active. Both mutants retained the ability to activate expression of the cyclin-dependent kinase inhibitor p2lcip1/waf1, and this activity correlated with the ability to induce a G1 cell cycle arrest. However, an extension of this survey to include other p53 targets showed that p53 175P was defective in the activation of p53-responsive sequences derived from the bax promoter and the insulin-like growth factor-binding protein 3 gene (IGF-BP3) promoter, while p53 181L showed loss of the ability to activate a promoter containing IGF-BP3 box B sequences. Failure to activate transcription was also reflected in the reduced ability of the mutants to bind the p53-responsive DNA sequences present in these promoters. These specific defects in transcriptional activation correlated with the impaired apoptotic function displayed by these mutants, and the results suggest that activation of cell cycle arrest genes by p53 can be separated from activation of genes with a role in mediating the p53 apoptotic response. The cellular response to p53 activation may therefore depend, at least in part, on which group of p53-responsive genes become transcriptionally activated.


Subject(s)
Apoptosis/physiology , Gene Expression Regulation , Genes, p53 , Promoter Regions, Genetic , Proto-Oncogene Proteins c-bcl-2 , Tumor Suppressor Protein p53/physiology , Cell Cycle/genetics , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , DNA/metabolism , Enzyme Activation , Humans , Neoplasm Proteins/metabolism , Neoplasms/genetics , Neoplasms/pathology , Point Mutation , Proto-Oncogene Proteins/metabolism , Transfection , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , bcl-2-Associated X Protein
11.
Mol Cell Biol ; 19(3): 1751-8, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10022862

ABSTRACT

The p53 tumor suppressor protein can be phosphorylated at several sites within the N- and C-terminal domains, and several protein kinases have been shown to phosphorylate p53 in vitro. In this study, we examined the activity of p53 proteins with combined mutations at all of the reported N-terminal phosphorylation sites (p53N-term), all of the C-terminal phosphorylation sites (p53C-term), or all of the phosphorylation sites together (p53N/C-term). Each of these mutant proteins retained transcriptional transactivation functions, indicating that phosphorylation is not essential for this activity of p53, although a subtle contribution of the C-terminal phosphorylation sites to the activation of expression of the endogenous p21(Waf1/Cip1)-encoding gene was detected. Mutation of the phosphorylation sites to alanine did not affect the sensitivity of p53 to binding to or degradation by Mdm2, although alteration of residues 15 and 37 to aspartic acid, which could mimic phosphorylation, resulted in a slight resistance to Mdm2-mediated degradation, consistent with recent reports that phosphorylation at these sites inhibits the p53-Mdm2 interaction. However, expression of the phosphorylation site mutant proteins in both wild-type p53-expressing and p53-null lines showed that all of the mutant proteins retained the ability to be stabilized following DNA damage. This indicates that phosphorylation is not essential for DNA damage-induced stabilization of p53, although phosphorylation could clearly contribute to p53 stabilization under some conditions.


Subject(s)
Tumor Suppressor Protein p53/metabolism , Binding Sites , Humans , Mutagenesis, Site-Directed , Phosphorylation , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
12.
Mol Cell Biol ; 18(10): 5690-8, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9742086

ABSTRACT

The stability of the p53 tumor suppressor protein is regulated by interaction with Mdm2, the product of a p53-inducible gene. Mdm2-targeted degradation of p53 depends on the interaction between the two proteins and is mediated by the proteasome. We show here that in addition to the N-terminal Mdm2 binding domain, the C terminus of p53 participates in the ability of p53 to be degraded by Mdm2. In contrast, alterations in the central DNA binding domain of p53, which change the conformation of the p53 protein, do not abrogate the sensitivity of the protein to Mdm2-mediated degradation. The importance of the C-terminal oligomerization domain to Mdm2-targeted degradation of p53 is likely to reflect the importance of oligomerization of the full-length p53 protein for interaction with Mdm2, as previously shown in vitro. Interestingly, the extreme C-terminal region of p53, outside the oligomerization domain, was also shown to be necessary for efficient degradation, and deletion of this region stabilized the protein without abrogating its ability to bind to Mdm2. Mdm2-resistant p53 mutants were not further stabilized following DNA damage, supporting a role for Mdm2 as the principal regulator of p53 stability in cells. The extreme C terminus of the p53 protein has previously been shown to contain several regulatory elements, raising the possibility that either allosteric regulation of p53 by this domain or interaction between this region and a third protein plays a role in determining the sensitivity of p53 to Mdm2-directed degradation.


Subject(s)
Nuclear Proteins , Proto-Oncogene Proteins/metabolism , Repressor Proteins , Tumor Suppressor Protein p53/metabolism , Animals , Cell Nucleus/metabolism , DNA Damage , Humans , Mice , Oncogene Proteins, Viral/metabolism , Proto-Oncogene Proteins c-mdm2 , Sequence Deletion , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
13.
Mol Cell Biol ; 21(24): 8521-32, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11713287

ABSTRACT

The growth inhibitory functions of p53 are controlled in unstressed cells by rapid degradation of the p53 protein. One of the principal regulators of p53 stability is MDM2, a RING finger protein that functions as an E3 ligase to ubiquitinate p53. MDM2 promotes p53 nuclear export, and in this study, we show that ubiquitination of the C terminus of p53 by MDM2 contributes to the efficient export of p53 from the nucleus to the cytoplasm. In contrast, MDM2 did not promote nuclear export of the p53-related protein, p73. p53 nuclear export was enhanced by overexpression of the export receptor CRM1, although no significant relocalization of MDM2 was seen in response to CRM1. However, nuclear export driven by CRM1 overexpression did not result in the degradation of p53, and nuclear export was not essential for p53 degradation. These results indicate that MDM2 mediated ubiquitination of p53 contributes to both nuclear export and degradation of p53 but that these activities are not absolutely dependent on each other.


Subject(s)
Active Transport, Cell Nucleus , Nuclear Proteins , Receptors, Cytoplasmic and Nuclear , Tumor Suppressor Protein p53/chemistry , Blotting, Western , Cell Division , Cell Line , Cell Nucleus/metabolism , Cells, Cultured , Cytoplasm/metabolism , Dose-Response Relationship, Drug , Humans , Karyopherins/metabolism , Lysine/chemistry , Microscopy, Fluorescence , Models, Genetic , Mutation , Plasmids/metabolism , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2 , Transfection , Ubiquitin/metabolism , Exportin 1 Protein
14.
Cell Death Differ ; 12(4): 369-76, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15731768

ABSTRACT

The E2F family of transcription factors regulates the expression of a number of genes whose products are involved in cell cycle control, DNA replication and apoptosis. We show here that E2F-1 binds in vivo the promoters of ASPP1 and ASPP2 genes, two activators of p53-mediated apoptosis, E2F-1, E2F-2 and E2F-3 all activate the isolated ASPP1 and ASPP2 promoters. Overexpression or deregulation of E2F-1 increased the expression levels of ASPP1 and ASPP2 mRNA and proteins. The identification of ASPP1 and ASPP2 genes as transcriptional targets of E2F provides another mechanism by which E2F cooperates with p53 to induce apoptosis.


Subject(s)
Carrier Proteins/genetics , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing , Apoptosis Regulatory Proteins , Base Sequence , Carrier Proteins/biosynthesis , E2F Transcription Factors , E2F1 Transcription Factor , E2F2 Transcription Factor , E2F3 Transcription Factor , Humans , Molecular Sequence Data , Promoter Regions, Genetic , RNA, Messenger/metabolism
17.
Cancer Res ; 59(11): 2516-21, 1999 Jun 01.
Article in English | MEDLINE | ID: mdl-10363964

ABSTRACT

The INK4A locus on human chromosome 9p21 encodes two genes that have been implicated in replicative senescence and tumor suppression, p16INK4A and p14ARF. In contrast to p16INK4A, which is up-regulated to high levels, we were unable to detect p14ARF protein in senescent human keratinocytes. Also, p53, an established target of p14ARF, did not increase, suggesting that p14ARF is not instrumental in human keratinocyte senescence. In neoplastic keratinocyte cultures, p16INK4A inactivation was invariably associated with the immortal phenotype, and there was evidence for the inactivation of p16INK4A, independent of p14ARF, in 6 of 10 lines that lacked large homozygous deletions. In contrast, we failed to detect exon 1beta mutations or p16INK4A-independent deletions. These results emphasize the previously proposed role for p16INK4A in human keratinocyte senescence but do not rule out a supporting role for p14ARF inactivation.


Subject(s)
Cellular Senescence/physiology , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Gene Deletion , Keratinocytes/physiology , Proteins/physiology , 3T3 Cells , Animals , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Line, Transformed , Cellular Senescence/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , Exons/genetics , Humans , Mice , Proteins/genetics , Tumor Cells, Cultured , Tumor Suppressor Protein p14ARF , Tumor Suppressor Protein p53/metabolism
18.
Cancer Res ; 56(22): 5285-92, 1996 Nov 15.
Article in English | MEDLINE | ID: mdl-8912870

ABSTRACT

The human papillomavirus type 16 (HPV-16), the type most often associated with cervical cancer, immortalizes primary keratinocytes and inhibits serum/calcium-stimulated differentiation in culture. In this study, we have used a model of keratinocyte immortalization based upon HPV-16 to analyze perturbation of function and expression of E-cadherin, a Ca(2+)-dependent cell-cell adhesion molecule expressed by normal keratinocytes, and its associated proteins. An immortalized keratinocyte cell line generated by cotransfection with HPV-16 E6 and E7 showed decreased membrane E-cadherin expression and redistribution of alpha-, beta-, and gamma-catenin from the undercoat membrane to the cytoplasm. No changes in the level of expression were seen. Selection of the immortalized keratinocyte cell line for resistance to differentiation generated a more transformed cell line with an invasive phenotype, down-regulated E-cadherin and alpha-catenin, and up-regulated the epidermal growth factor receptor (EGFr). Transfection of an E-cadherin expression construct into the differentiation-resistant cell line restored membrane-bound E-cadherin and catenin expression, down-regulated the EGFr, and reversed the invasive phenotype. These results indicate that overexpression of the EGFr correlates with perturbation of the E-cadherin/catenin complex seen in the HPV-16 E6- and E7-transfected keratinocytes and may underlie a functional interaction between growth-regulatory factors and adhesion molecules (E-cadherin/catenin).


Subject(s)
Cadherins/physiology , ErbB Receptors/physiology , Keratinocytes/metabolism , Oncogene Proteins, Viral/physiology , Proto-Oncogene Proteins/physiology , Repressor Proteins , Animals , Cadherins/genetics , Cadherins/metabolism , Cell Differentiation , Cell Line, Transformed , Down-Regulation , ErbB Receptors/metabolism , Humans , Keratinocytes/virology , Mice , Mice, Nude , Neoplasm Invasiveness , Oncogene Proteins, Viral/metabolism , Papillomaviridae , Papillomavirus E7 Proteins , Phenotype , Proto-Oncogene Proteins/metabolism , Receptor, ErbB-2/metabolism , Receptor, ErbB-2/physiology , Transfection , Up-Regulation
19.
Oncogene ; 4(2): 153-8, 1989 Feb.
Article in English | MEDLINE | ID: mdl-2538790

ABSTRACT

We have analysed the immortalizing function of Human Papillomavirus type 16 (HPV16) in a rat cell line which has been derived by immortalizing rat embryo fibroblasts with a thermolabile SV40 large T antigen and is temperature sensitive for growth. Introduction of wild type SV40 large T antigen or the adenovirus E1a 12s gene product has previously been shown to readily overcome the inability of these cells to divide at the non-permissive temperature. In contrast, the introduction of myc, another known immortalizing gene, cannot complement the growth defect of these cells. This cell line therefore provides a novel assay system which can distinguish two groups of immortalizing oncogenes. We have shown here that expression of HPV16 can readily complement the growth defect in this rat cell line and that this function can be genetically localised to E7. Cells expressing HPV16 E6 sequences are also weakly rescued from growth arrest at the non-permissive temperature. These results demonstrate a functional similarity between HPV16 E7, SV40 large T and adenovirus E1a and suggest that these genes may immortalize cells by a common mechanism. Interestingly, the limited sequence homology between these three gene products is restricted to the domain which has recently been implicated in binding the retinoblastoma gene product.


Subject(s)
Antigens, Polyomavirus Transforming/analysis , Cell Transformation, Viral , Oncogene Proteins, Viral/analysis , Papillomaviridae/physiology , Adenovirus Early Proteins , Amino Acid Sequence , Animals , Cell Line , Proto-Oncogenes , Rats , Retinoblastoma/genetics , Transfection
20.
Oncogene ; 18(53): 7637-43, 1999 Dec 13.
Article in English | MEDLINE | ID: mdl-10618703

ABSTRACT

Leading the way in imposing a policy of zero tolerance of cellular abnormalities that might lead to tumor development is the p53 protein. The efficiency of p53 in preventing cell growth is a strong deterrent to malignant progression, but this activity must be kept tightly restrained to allow normal cell growth and development. Essential components of this regulation are the mechanisms by which the p53 protein is degraded, and efficient turnover of p53 in normal cells prevents the accumulation of the protein. Modulation of these degradation pathways in response to stress leads to the rapid stabilization and accumulation of p53, and activation of the p53 response. It is now becoming clear that the Mdm2 protein is central to the regulation of p53 stability and multiple pathways exist through which the activity of Mdm2 can be inhibited. Defects in the ability to stabilize p53 are likely to contribute to malignant development, and restoration of this activity represents an extremely attractive possibility for tumor therapy.


Subject(s)
Nuclear Proteins , Tumor Suppressor Protein p53/metabolism , Animals , Humans , Oncogenes/genetics , Oncogenes/physiology , Phosphorylation , Proteins/physiology , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-mdm2 , Tumor Suppressor Protein p14ARF
SELECTION OF CITATIONS
SEARCH DETAIL