Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 178
Filter
1.
J Virol ; 75(24): 12347-58, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11711625

ABSTRACT

All members of the herpesvirus family persist in their host throughout life. In doing so, herpesviruses exploit a surprising number of different strategies to evade the immune system. Human herpesvirus 7 (HHV-7) is a relatively recently discovered member of the herpesvirus family, and little is known about how it escapes immune detection. Here we show that HHV-7 infection results in premature degradation of major histocompatibility complex class I molecules. We identify and characterize a protein from HHV-7, U21, that binds to and diverts properly folded class I molecules to a lysosomal compartment. Thus, U21 is likely to function in the normal course of HHV-7 infection to downregulate surface class I molecules and prevent recognition of infected cells by cytotoxic T lymphocytes.


Subject(s)
Glycoproteins/physiology , Herpesvirus 7, Human/physiology , Histocompatibility Antigens Class I/metabolism , Lysosomes/metabolism , Viral Proteins/physiology , Amino Acid Sequence , Cell Line , Concanavalin A/pharmacology , Genes, MHC Class I , Glycoproteins/chemistry , Glycoproteins/genetics , Histocompatibility Antigens Class I/chemistry , Humans , Molecular Sequence Data , Molecular Weight , Protein Folding
2.
Mol Cell Biol ; 21(13): 4169-76, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11390646

ABSTRACT

Virus infection of numerous cell types results in the transcriptional induction of a subset of virus- and interferon (IFN)-stimulated genes. The beta IFN (IFN-beta) gene is one of these rapidly induced genes; it serves as a fundamental component of the cellular defense response in eliciting potent antiviral, immunomodulatory, and antiproliferative effects. One of the transcription factors involved in the stringent regulation of IFN-beta production following virus infection is interferon regulatory factor (IRF) 3 (IRF-3). We have characterized an alternatively spliced isoform of IRF-3 that we have called IRF-3a. IRF-3a can selectively and potently inhibit virus-induced activation of the IFN-beta promoter. IRF-3a lacks half of the DNA binding domain found in IRF-3 and is unable to bind to the classical IRF binding elements, IFN-stimulated response elements. These studies suggest that IRF-3a may act as a modulator of IRF-3.


Subject(s)
Alternative Splicing , DNA-Binding Proteins/genetics , Gene Expression Regulation , Interferon-beta/genetics , Regulatory Sequences, Nucleic Acid/genetics , Transcription Factors/genetics , Amino Acid Sequence , Base Sequence , Cell Line , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Dimerization , Humans , Immunoblotting , Interferon Regulatory Factor-3 , Interferon-beta/metabolism , Molecular Sequence Data , Precipitin Tests , Promoter Regions, Genetic , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Structure, Tertiary , Respirovirus/physiology , Transcription Factors/chemistry , Transcription Factors/metabolism
3.
Genes Dev ; 14(22): 2813-8, 2000 Nov 15.
Article in English | MEDLINE | ID: mdl-11090129

ABSTRACT

Interferon regulatory factors constitute a family of transcriptional activators and repressors involved in a large number of vital cellular processes. Interferon regulatory factor-3 (IRF-3) has been implicated in virus and double-stranded RNA mediated induction of IFNbeta and RANTES, in DNA damage signaling, and in virus-induced apoptosis. With its critical role in these pathways, the activity of IRF-3 is tightly regulated in myriad ways. Here we describe novel regulation of IRF-3 at the level of RNA splicing. We show that an unprecedented dual utilization of a splice acceptor/donor site within the IRF-3 mRNA governs the production of two alternative splice isoforms.


Subject(s)
Alternative Splicing , DNA-Binding Proteins/genetics , Transcription Factors/genetics , Amino Acid Sequence , Base Sequence , Cells, Cultured , Genomic Library , Humans , Interferon Regulatory Factor-3 , Molecular Sequence Data , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Protein Binding , RNA-Binding Proteins/metabolism , Serine-Arginine Splicing Factors
4.
EMBO J ; 19(21): 5762-71, 2000 Nov 01.
Article in English | MEDLINE | ID: mdl-11060027

ABSTRACT

A hallmark of human papillomavirus (HPV) associated carcinogenesis is the integration of the viral DNA into the cellular genome, usually accompanied by the loss of expression of the viral E2 gene. E2 binds to and represses the viral promoter directing expression of the E6 and E7 oncogenes. The re-introduction and expression of exogenous E2 in HPV-positive cancer cells results in cellular growth arrest, while growth in the context of exogenous E2 can be restored through the expression of exogenous E6 and E7. Here we examine the individual contributions of the viral E6 and E7 genes to this phenotype. E6 alone displays moderate activity, whereas both E7 and adenovirus E1A display high activity in reversing E2-mediated cellular growth suppression. Using defined mutants of E7 and E1A, we show that an intact retinoblastoma interaction domain is required for this function. In addition, we show that the E2-mediated growth arrest of HPV-positive cells results in cellular senescence, and implicate the cyclin/cdk inhibitor p21(CIP) as a downstream E2 effector in this phenotype.


Subject(s)
Cellular Senescence/physiology , DNA-Binding Proteins , Genes, Viral , Papillomaviridae/genetics , Papillomaviridae/physiology , Repressor Proteins , Base Sequence , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , Cyclins/physiology , Female , HeLa Cells , Humans , Oligodeoxyribonucleotides, Antisense/genetics , Oligodeoxyribonucleotides, Antisense/pharmacology , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/physiology , Papillomaviridae/pathogenicity , Papillomavirus E7 Proteins , Phenotype , Retinoblastoma Protein/genetics , Retinoblastoma Protein/physiology , Transfection , Tumor Cells, Cultured , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/virology
5.
Mol Cell ; 6(3): 751-6, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11030355

ABSTRACT

The functional characterization of a specific gene, or its protein product, often relies on assessing the consequences of its elimination, usually accomplished by gene knockout, ribozyme, antisense, or RNA-mediated interference (RNAi) technologies. The selective degradation of cellular proteins is mediated primarily by the ubiquitin-proteasome pathway. Manipulation of the ubiquitin-dependent proteolytic machinery to eliminate specific gene products at the protein level has been previously attempted with some success in vitro; however, the in vivo efficacy of this approach has not yet been achieved. Here we report successful engineering of the substrate receptor of a major ubiquitin-proteolytic machinery to direct the degradation of otherwise stable cellular proteins both in yeast and in mammalian cells.


Subject(s)
F-Box Proteins , Ligases/genetics , Molecular Biology/methods , Retinoblastoma Protein/metabolism , Ubiquitins/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , F-Box-WD Repeat-Containing Protein 7 , Female , GTP-Binding Proteins/metabolism , Gene Expression Regulation, Enzymologic , Humans , Ligases/metabolism , Mammals , Nuclear Proteins/metabolism , Osteosarcoma , Plasmids , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Retinoblastoma-Like Protein p107 , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins , Substrate Specificity/genetics , Tumor Cells, Cultured , Ubiquitin-Protein Ligases , Uterine Cervical Neoplasms , beta-Transducin Repeat-Containing Proteins
6.
Mol Cell ; 6(2): 409-19, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10983987

ABSTRACT

Although there is a binding site on the proteasome for the polyubiquitin chains attached to degradation substrates by the ubiquitination machinery, it is currently unclear whether in vivo the activities of the ubiquitination machinery and the proteasome are coupled. Here we show that two human homologs of the yeast ubiquitin-like Dsk2 protein, hPLIC-1 and hPLIC-2, physically associate with both proteasomes and ubiquitin ligases in large complexes. Overexpression of hPLIC proteins interferes with the in vivo degradation of two unrelated ubiquitin-dependent proteasome substrates, p53 and IkappaBalpha, but not a ubiquitin-independent substrate. Our findings raise the possibility that the hPLIC proteins, and possibly related ubiquitin-like family members, may functionally link the ubiquitination machinery to the proteasome to affect in vivo protein degradation.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , Cysteine Endopeptidases/metabolism , Multienzyme Complexes/metabolism , Saccharomyces cerevisiae Proteins , Ubiquitins/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Animals , Autophagy-Related Proteins , Cells, Cultured , Fungal Proteins/chemistry , Fungal Proteins/metabolism , Gene Expression Regulation/drug effects , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Male , Mice , Molecular Sequence Data , Proteasome Endopeptidase Complex , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Skin/cytology , Transfection , Tumor Necrosis Factor-alpha/pharmacology , Ubiquitins/chemistry , Ubiquitins/genetics
7.
J Virol ; 74(14): 6408-17, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10864652

ABSTRACT

The E6 protein of the high-risk human papillomaviruses (HPVs) and the cellular ubiquitin-protein ligase E6AP form a complex which causes the ubiquitination and degradation of p53. We show here that HPV16 E6 promotes the ubiquitination and degradation of E6AP itself. The half-life of E6AP is shorter in HPV-positive cervical cancer cells than in HPV-negative cervical cancer cells, and E6AP is stabilized in HPV-positive cancer cells when expression of the viral oncoproteins is repressed. Expression of HPV16 E6 in cells results in a threefold decrease in the half-life of transfected E6AP. E6-mediated degradation of E6AP requires (i) the binding of E6 to E6AP, (ii) the catalytic activity of E6AP, and (iii) activity of the 26S proteasome, suggesting that E6-E6AP interaction results in E6AP self-ubiquitination and degradation. In addition, both in vitro and in vivo experiments indicate that E6AP self-ubiquitination results primarily from an intramolecular transfer of ubiquitin from the active-site cysteine to one or more lysine residues; however, intermolecular transfer can also occur in the context of an E6-mediated E6AP multimer. Finally, we demonstrate that an E6 mutant that is able to immortalize human mammary epithelial cells but is unable to degrade p53 retains its ability to bind and degrade E6AP, raising the possibility that E6-mediated degradation of E6AP contributes to its ability to transform mammalian cells.


Subject(s)
Ligases/metabolism , Oncogene Proteins, Viral/metabolism , Papillomaviridae/metabolism , Proteasome Endopeptidase Complex , Repressor Proteins , Ubiquitins/metabolism , Blotting, Western , Catalysis , Humans , Mutation , Oncogene Proteins, Viral/genetics , Peptide Hydrolases/metabolism , Polymerase Chain Reaction , Transfection , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases
8.
J Virol ; 74(8): 3752-60, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10729150

ABSTRACT

The papillomavirus E2 gene product plays a pivotal role in viral replication. E2 has multiple functions, including (i) transcriptional activation and repression of viral promoters and (ii) the enhancement of viral DNA replication. It was previously reported that E2 suppressed the growth of papillomavirus-positive cervical carcinoma cell lines. In the present study, we investigated the mechanisms of E2 growth inhibition. We found that the transcriptional activation function of E2 is required for inhibition of the growth of HeLa cells as well as for transcriptional repression of the viral E6/E7 promoter. It had been previously postulated that transcriptional repression of the E6/E7 promoter results from E2 binding its cognate sites proximal to the E6/E7 promoter and displacing other cellular transcriptional factors. In this study, we report a requirement for the transcription activation function for the binding of E2 to transcriptionally active templates.


Subject(s)
Cell Division , DNA-Binding Proteins , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/physiology , Papillomaviridae/genetics , Transcriptional Activation , DNA Replication , DNA, Viral/metabolism , Gene Expression Regulation, Viral , HeLa Cells , Humans , Oncogene Proteins, Viral/metabolism , Papillomaviridae/physiology , Papillomavirus E7 Proteins , Plasmids/genetics , Promoter Regions, Genetic , Repressor Proteins/genetics , Repressor Proteins/metabolism , Templates, Genetic , Transcription, Genetic , Transfection
9.
J Virol ; 74(6): 2679-86, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10684283

ABSTRACT

The human papillomavirus (HPV) E2 protein is an important regulator of viral E6 and E7 gene expression. E2 can repress the viral promoter for E6 and E7 expression as well as block progression of the cell cycle in cancer cells harboring the DNA of "high-risk" HPV types. Although the phenomenon of E2-mediated growth arrest of HeLa cells and other HPV-positive cancer cells has been well documented, the specific mechanism by which E2 affects cellular proliferation has not yet been elucidated. Here, we show that bovine papillomavirus (BPV) E2-induced growth arrest of HeLa cells requires the repression of the E6 and E7 promoter. This repression is specific for E2TA and not E2TR, a BPV E2 variant that lacks the N-terminal transactivation domain. We demonstrate that expression of HPV16 E6 and E7 from a heterologous promoter that is not regulated by E2 rescues HeLa cells from E2-mediated growth arrest. Our data indicate that the pathway of E2-mediated growth arrest of HeLa cells requires repression of E6 and E7 expression through an activity specified by the transactivation domain of E2TA.


Subject(s)
Gene Expression Regulation, Viral , Oncogene Proteins, Viral/genetics , Papillomaviridae/genetics , Promoter Regions, Genetic , Repressor Proteins , Uterine Cervical Neoplasms/virology , Virus Integration , Animals , Cattle , Cell Division , DNA-Binding Proteins/genetics , Female , Humans , Papillomavirus E7 Proteins , Tumor Cells, Cultured , Viral Proteins/genetics
10.
Science ; 286(5443): 1321-6, 1999 Nov 12.
Article in English | MEDLINE | ID: mdl-10558980

ABSTRACT

The E6AP ubiquitin-protein ligase (E3) mediates the human papillomavirus-induced degradation of the p53 tumor suppressor in cervical cancer and is mutated in Angelman syndrome, a neurological disorder. The crystal structure of the catalytic hect domain of E6AP reveals a bilobal structure with a broad catalytic cleft at the junction of the two lobes. The cleft consists of conserved residues whose mutation interferes with ubiquitin-thioester bond formation and is the site of Angelman syndrome mutations. The crystal structure of the E6AP hect domain bound to the UbcH7 ubiquitin-conjugating enzyme (E2) reveals the determinants of E2-E3 specificity and provides insights into the transfer of ubiquitin from the E2 to the E3.


Subject(s)
Ligases/chemistry , Ligases/metabolism , Ubiquitins/metabolism , Amino Acid Sequence , Angelman Syndrome/genetics , Binding Sites , Catalytic Domain , Conserved Sequence , Crystallography, X-Ray , Cysteine/chemistry , Humans , Models, Molecular , Molecular Sequence Data , Mutation , Protein Conformation , Protein Structure, Secondary , Substrate Specificity , Ubiquitin-Conjugating Enzymes , Ubiquitin-Protein Ligases
11.
Proc Natl Acad Sci U S A ; 96(24): 13738-43, 1999 Nov 23.
Article in English | MEDLINE | ID: mdl-10570142

ABSTRACT

Src family tyrosine kinases are involved in modulating various signal transduction pathways leading to the induction of DNA synthesis and cytoskeletal reorganization in response to cell-cell or cell-matrix adhesion. The critical role of these kinases in regulating cellular signaling pathways requires that their activity be tightly controlled. Src family proteins are regulated through reversible phosphorylation and dephosphorylation events that alter the conformation of the kinase. We have found evidence that Src also is regulated by ubiquitination. Activated forms of Src are less stable than either wild-type or kinase-inactive Src mutants and can be stabilized by proteasome inhibitors. In addition, poly-ubiquitinated forms of active Src have been detected in vivo. Taken together, our results establish ubiquitin-mediated proteolysis as a previously unidentified mechanism for irreversibly attenuating the effects of active Src kinase.


Subject(s)
Ubiquitins/metabolism , src-Family Kinases/metabolism , Animals , COS Cells , CSK Tyrosine-Protein Kinase , Chickens , Enzyme Activation , Oncogene Protein pp60(v-src)/genetics , Oncogene Protein pp60(v-src)/metabolism , Protein-Tyrosine Kinases , Proto-Oncogene Proteins pp60(c-src)/genetics , Proto-Oncogene Proteins pp60(c-src)/metabolism , src-Family Kinases/genetics
12.
Proc Natl Acad Sci U S A ; 96(17): 9557-62, 1999 Aug 17.
Article in English | MEDLINE | ID: mdl-10449731

ABSTRACT

The Src family of nonreceptor tyrosine kinases are important regulators of a variety of cellular processes, including cytoskeletal organization, cell-cell contact, and cell-matrix adhesion. Activation of Src family kinases also can induce DNA synthesis and cellular proliferation; therefore, tight regulation of their kinase activities is important for the cell to maintain proliferative control. Posttranslational phosphorylation and dephosphorylation are recognized as the principle modifications by which the activities of the Src family of tyrosine kinases are regulated. We have discovered that this family of kinases also is regulated by ubiquitin-mediated proteolysis. Studies aimed at the identification of cellular targets for E6AP, an E3 ubiquitin protein ligase involved in ubquitin-mediated degradation, led us to the identification of members of the Src family kinases as potential substrates for E6AP. We have found that E6AP can bind to several of the Src family tyrosine kinases. Here we show that activated Blk is preferentially degraded by the ubiquitin-proteasome pathway and that its ubiquitination is mediated by E6AP. Identification of members of the Src tyrosine kinase family as substrates of the E6AP ubiquitin-protein ligase implicates a role for the ubiquitin pathway in regulating the activities of individual members of this important family of signaling molecules.


Subject(s)
Ligases/metabolism , Ubiquitins/metabolism , src-Family Kinases/metabolism , Animals , Blotting, Western , COS Cells , Cells, Cultured , Enzyme Activation , Enzyme Stability , Mice , Transfection , Ubiquitin-Protein Ligases
13.
J Biol Chem ; 274(26): 18785-92, 1999 Jun 25.
Article in English | MEDLINE | ID: mdl-10373495

ABSTRACT

The human papilloma virus E6-associated protein (E6AP) functions as a ubiquitin protein ligase (E3) in the E6-mediated ubiquitination of p53. E6AP is also an E3 in the absence of E6, but its normal cellular substrates have not yet been identified. Here we report the identification of HHR23A, one of the human homologues of the yeast DNA repair protein Rad23, as an E6-independent target of E6AP. HHR23A binds E6AP and is ubiquitinated in vitro in an E6AP-dependent manner. Ubiquitinated forms of endogenous HHR23A are detectable in mammalian cells. Overexpression of wild-type E6AP in vivo enhances the ubiquitination of HHR23A, whereas a dominant negative E6AP mutant inhibits HHR23A ubiquitination. Although HHR23A is a stable protein in non-synchronized cells, its levels are regulated in a cell cycle-dependent manner, with specific degradation occurring during S phase. The S phase degradation of HHR23A could be blocked in vivo by dominant negative E6AP, providing direct evidence for the involvement of E6AP in the regulation of HHR23A. Consistent with a role of the HHR23 proteins in DNA repair, UV-induced DNA damage inhibited HHR23A degradation. Although the precise role of HHR23 proteins in DNA repair and cell cycle progression remains to be elucidated, our data suggest that E6AP-mediated ubiquitination of HHR23A may have important implications in DNA repair and cell cycle progression.


Subject(s)
DNA Repair , DNA-Binding Proteins/metabolism , Ligases/metabolism , Ubiquitins/metabolism , Cell Cycle , DNA Repair Enzymes , Humans , Ligases/genetics , Mutagenesis, Site-Directed , Transfection , Ubiquitin-Protein Ligases , Ultraviolet Rays , Xeroderma Pigmentosum/genetics , Xeroderma Pigmentosum/metabolism
14.
Oncogene ; 18(21): 3205-12, 1999 May 27.
Article in English | MEDLINE | ID: mdl-10359526

ABSTRACT

Phosphorylation of the p53 tumor suppressor protein is likely to play an important role in regulating its activity. To study the regulatory role of potential phosphorylation sites within the N-terminal transactivation domain of human p53 (hp53), a series of p53 serine mutants were evaluated for transcriptional transactivation and sequence specific DNA binding. The role of these mutations in regulating p53-mediated growth suppression and programmed cell death was examined. This mutational analysis comprised serine residues located at positions 6, 9, 15, 20, 33 and 37 of human p53. Substitution of serine for alanine, either at individual residues or at all six residues together, did not affect the suppression of cell growth and cell transformation, or the ability to bind DNA specifically and to transactivate different promoters, nor did it alter p53 expression. However, the ability of p53 to induce apoptosis was impaired by specific serine substitutions. Mutations in all six N-terminal serines together reduced the apoptotic activity of p53 in H1299 cells by 50%. Analysis of individual mutants revealed that mutations in serine 15 and 20 are primarily responsible for this impairment. Our results suggest that these serines play a role in the regulation of p53-mediated apoptosis.


Subject(s)
Apoptosis , Mutagenesis, Site-Directed , Serine/metabolism , Tumor Suppressor Protein p53/metabolism , Alanine/genetics , Alanine/metabolism , Animals , Binding Sites , Cell Transformation, Neoplastic , DNA/metabolism , Gene Expression , Humans , Mice , Serine/genetics , Transcription, Genetic , Transcriptional Activation , Tumor Suppressor Protein p53/genetics
15.
J Virol Methods ; 79(1): 65-74, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10328536

ABSTRACT

Measles virus encodes three proteins required for the encapsidation, transcription and replication of viral genomes. The genes for these proteins have been inserted into the vaccinia virus genome together with the gene for the bacteriophage T7 RNA polymerase. Cells infected with this recombinant virus were able to encapsidate, transcribe and replicate a CAT gene positioned in the negative polarity behind a T7 promoter and flanked by measles virus genomic termini. Inhibition of the accumulation of the nucleocapsid proteins by actinomycin D led to an increase in CAT expression. Thus the measles virus polymerase activity, encoded by the vaccinia genome, was regulated by the level of measles proteins just as the authentic polymerase. The recombinant vaccinia described in this study could be useful for the production of measles virus-like particles encoding foreign genes and employed in vaccination or gene therapy strategies.


Subject(s)
Measles virus/genetics , Measles virus/physiology , Transcription, Genetic/genetics , Vaccinia virus/genetics , Virus Replication , Bacteriophage T7/enzymology , Chloramphenicol O-Acetyltransferase/genetics , Chloramphenicol O-Acetyltransferase/metabolism , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Genetic Vectors , Genome, Viral , Nucleocapsid/genetics , Nucleocapsid/metabolism , Recombination, Genetic , Restriction Mapping , Reverse Transcriptase Polymerase Chain Reaction
16.
Mol Cell ; 2(5): 571-80, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9844630

ABSTRACT

The S. cerevisiae SCFCdc4p ubiquitin-protein ligase complex promotes cell cycle transitions through degradation of cell cycle regulators. To investigate SCFCdc4p regulation in vivo, we examined the stability of individual SCFCdc4p components. Whereas Cdc53p and Skp1p were stable, Cdc4p, the F box-containing component responsible for substrate recognition, was short lived and subject to SCF-mediated ubiquitination. Grr1p, another F box component of SCF complexes, was also ubiquitinated. A stable truncated Cdc4pF-beta-gal hybrid protein capable of binding Skp1p and entering into an SCF complex interfered with proteolysis of SCF targets and inhibited cell proliferation. The finding that the F box-containing SCF components are unstable suggests a mechanism of regulating SCF function through ubiquitination and proteolysis of F box components.


Subject(s)
Carrier Proteins , Cell Cycle Proteins/metabolism , Cullin Proteins , F-Box Proteins , Ligases/metabolism , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/metabolism , Ubiquitin-Protein Ligase Complexes , Anaphase-Promoting Complex-Cyclosome , CDC28 Protein Kinase, S cerevisiae/genetics , CDC28 Protein Kinase, S cerevisiae/physiology , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Cycle Proteins/genetics , Cyclins/metabolism , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Fungal Proteins/genetics , Fungal Proteins/metabolism , Half-Life , Hydroxyurea/pharmacology , Ligases/genetics , Ligases/physiology , Lipoproteins/pharmacology , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Mutation , Nocodazole/pharmacology , Pheromones , Precipitin Tests , Proteasome Endopeptidase Complex , Recombinant Fusion Proteins/metabolism , S-Phase Kinase-Associated Proteins , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Substrate Specificity , Ubiquitin-Protein Ligases , Ubiquitins/metabolism
17.
EMBO J ; 17(22): 6714-22, 1998 Nov 16.
Article in English | MEDLINE | ID: mdl-9822614

ABSTRACT

The NOT genes were originally identified in a yeast genetic screen that selected mutations resulting in increased utilization of a non-consensus TC TATA element of the HIS3 promoter. Here, we present evidence that the N-terminus of Not2 interacts with components of the Ada/Gcn5 histone acetyltransferase complex. Loss of this interaction either through abrogation of Not2 N-terminal function or deletion of ada2 or gcn5 results in derepression of the HIS3 TC element. This suggests that association of Not2 with the Ada/Gcn5 histone acetyltransferase complex is involved in regulation of the HIS3 promoter. Association between the Not and CCR4 transcriptional regulatory complexes has also been observed recently. Our phenotypic analyses suggest that these CCR4-related Not2 functions are mediated by a functionally independent domain of Not2 that includes the highly conserved C-terminus. Chimeric proteins containing the yeast Not2 N-terminus fused to the human C-terminus function in yeast, suggesting that the Not2 C-terminus represents a distinct modular domain whose function is conserved between higher and lower eukaryotes.


Subject(s)
Acetyltransferases/metabolism , Cell Cycle Proteins/metabolism , DNA-Binding Proteins , Fungal Proteins/metabolism , Protein Kinases/metabolism , Repressor Proteins/metabolism , Ribonucleases , Saccharomyces cerevisiae Proteins , Transcription Factors/metabolism , Amino Acid Sequence , Bacterial Proteins/genetics , Cell Cycle Proteins/genetics , Fungal Proteins/genetics , Histone Acetyltransferases , Humans , Hydro-Lyases/genetics , Molecular Sequence Data , Promoter Regions, Genetic , Protein Binding , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Repressor Proteins/genetics , Serine Endopeptidases/genetics
18.
Mol Cell ; 2(4): 405-15, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9809062

ABSTRACT

Control of p53 turnover is critical to p53 function. E1A binding to p300/CBP translates into enhanced p53 stability, implying that these coactivator proteins normally operate in p53 turnover control. In this regard, the p300 C/H1 region serves as a specific in vivo binding site for both p53 and MDM2, a naturally occurring p53 destabilizer. Moreover, most of the endogenous MDM2 is bound to p300, and genetic analysis implies that specific interactions of p53 and MDM2 with p300 C/H1 are important steps in the MDM2-directed turnover of p53. A specific role for p300 in endogenous p53 degradation is underscored by the p53-stabilizing effect of overproducing the p300 C/H1 domain. Taken together, the data indicate that specific interactions between p300/CBP C/H1, p53, and MDM2 are intimately involved in the MDM2-mediated control of p53 abundance.


Subject(s)
DNA-Binding Proteins , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Tumor Suppressor Protein p53/metabolism , Cell Line , Humans , Molecular Sequence Data , Mutagenesis/physiology , Neoplasm Proteins/metabolism , Nuclear Proteins/genetics , Oncogene Proteins, Viral/genetics , Protein Binding/physiology , Proto-Oncogene Proteins c-mdm2 , Sequence Homology, Amino Acid , Trans-Activators/genetics , Ubiquitins/metabolism
19.
J Biol Chem ; 273(37): 24057-64, 1998 Sep 11.
Article in English | MEDLINE | ID: mdl-9727024

ABSTRACT

Cyclin-dependent kinase 5 (Cdk5) was originally isolated by its close homology to the human CDC2 gene, which is a key regulator of cell cycle progression. However, unlike other Cdks, the activity of Cdk5 is required in post-mitotic neurons. The neuronal-specific p35 protein, which shares no homology to cyclins, was identified by virtue of its association and activation of Cdk5. Gene targeting studies in mice have shown that the p35/Cdk5 kinase is required for the proper neuronal migration and development of the mammalian cortex. We have investigated the regulation of the p35/Cdk5 kinase. Here we show that p35, the activator of Cdk5, is a short-lived protein with a half-life (t1/2) of 20 to 30 min. Specific proteasome inhibitors such as lactacystin greatly stabilize p35 in vivo. Ubiquitination of p35 can be readily demonstrated in vitro and in vivo. Inhibition of Cdk5 activity by a specific Cdk inhibitor, roscovitine, or by overexpression of a dominant negative mutant of Cdk5 increases the stability of p35 by 2- to 3-fold. Furthermore, phosphorylation mutants of p35 also stabilize p35 2- to 3-fold. Together, these observations demonstrate that the p35/Cdk5 kinase can be subject to rapid turnover in vivo and suggest that phosphorylation of p35 upon Cdk5 kinase activation plays a autoregulatory role in p35 degradation mediated by ubiquitin-mediated proteolysis.


Subject(s)
Cyclin-Dependent Kinases , Cysteine Endopeptidases/metabolism , Multienzyme Complexes/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Protein Serine-Threonine Kinases/metabolism , Ubiquitins/metabolism , Acetylcysteine/analogs & derivatives , Acetylcysteine/pharmacology , Animals , COS Cells , Cells, Cultured , Cerebral Cortex/metabolism , Cyclin-Dependent Kinase 5 , Embryo, Mammalian , Enzyme Activation , Enzyme Inhibitors/pharmacology , Half-Life , Humans , Kinetics , Mammals , Mice , Nerve Tissue Proteins/genetics , Neurons/cytology , Polymerase Chain Reaction , Proteasome Endopeptidase Complex , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Purines/pharmacology , Rats , Rats, Sprague-Dawley , Recombinant Proteins/metabolism , Roscovitine , Sequence Deletion , Transfection
20.
J Virol ; 72(10): 8166-73, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9733858

ABSTRACT

Mutation of the conserved glutamic acid residue at position 39 of human papillomavirus type 16 (HPV-16) E2 to alanine (E39A) disrupts its E1 interaction activity and its replication function in transient replication assays but does not affect E2 transcriptional activation. This E39A mutation also disrupts replication activity of HPV-16 E2 in HPV-16 in vitro DNA replication. On this basis, we designed 23- and 15-amino-acid peptides derived from HPV-16 E2 sequences flanking the E39 residue and tested the ability of these peptides to inhibit interaction between HPV-16 E1 and E2 in vitro. The inhibitory activity of these peptides was specific, since analogous peptides in which alanine was substituted for the E39 residue did not inhibit interaction. The 15-amino-acid peptide E2N-WP15 was the smallest peptide tested that effectively inhibited HPV-16 E1-E2 interaction. This peptide also inhibited in vitro replication of HPV-16 DNA. The efficacy of E2N-WP15 was not exclusive to HPV-16: this peptide also inhibited interaction of HPV-11 E1 with the E2 proteins of both HPV-11 and HPV-16 and inhibited in vitro replication with these same combinations of E1 and E2 proteins. These results provide further evidence that E1-E2 interaction is required for papillomavirus DNA replication and constitute the first demonstration that inhibition of this interaction is sufficient to prevent HPV DNA replication in vitro.


Subject(s)
DNA Replication/drug effects , DNA-Binding Proteins , Oncogene Proteins, Viral/metabolism , Oncogene Proteins/metabolism , Papillomaviridae/genetics , Peptides/pharmacology , Amino Acid Sequence , DNA, Viral/biosynthesis , Molecular Sequence Data , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...