Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
J Virol ; 75(20): 9844-56, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11559818

ABSTRACT

A major impediment to successful chemotherapy is the propensity for some tumor cells to undergo cell cycle arrest rather than apoptosis. It is well established, however, that the adenovirus E1A protein can sensitize these cells to the induction of apoptosis by anticancer agents. To further understand how E1A enhances chemosensitivity, we have made use of a human colon carcinoma cell line (HCT116) which typically undergoes cell cycle arrest in response to chemotherapeutic drugs. As seen by the analysis of E1A mutants, we show here that E1A can induce apoptosis in these cells by neutralizing the activities of the cyclin-dependent kinase inhibitor p21. E1A's ability to interact with p21 and thereby restore Cdk2 activity in DNA-damaged cells correlates with the reversal of G(1) arrest, which in turn leads to apoptosis. Analysis of E1A mutants failing to bind p300 (also called CBP) or Rb shows that they are almost identical to wild-type E1A in their ability to initially overcome a G(1) arrest in cells after DNA damage, while an E1A mutant failing to bind p21 is not. However, over time, this mutant, which can still target Rb, is far more efficient in accumulating cells with a DNA content greater than 4N but is similar to wild-type E1A and the other E1A mutants in releasing cells from a p53-mediated G(2) block following chemotherapeutic treatment. Thus, we suggest that although E1A requires the binding of p21 to create an optimum environment for apoptosis to occur in DNA-damaged cells, E1A's involvement in other pathways may be contributing to this process as well. A model is proposed to explain the implications of these findings.


Subject(s)
Adenovirus E1A Proteins/pharmacology , Apoptosis , CDC2-CDC28 Kinases , DNA Damage , Protein Serine-Threonine Kinases/antagonists & inhibitors , Adenovirus E1A Proteins/genetics , Antineoplastic Agents/pharmacology , Cell Cycle , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinases/metabolism , Doxorubicin/pharmacology , G1 Phase , Humans , Mutation , Nuclear Proteins/physiology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Trans-Activators/physiology , Transfection , Tumor Cells, Cultured , p21-Activated Kinases
2.
EMBO J ; 20(7): 1739-53, 2001 Apr 02.
Article in English | MEDLINE | ID: mdl-11285237

ABSTRACT

The molecular mechanism(s) that are responsible for suppressing MyoD's transcriptional activities in undifferentiated skeletal muscle cells have not yet been determined. We now show that MyoD associates with a histone deacetylase-1 (HDAC1) in these cells and that this interaction is responsible for silencing MyoD-dependent transcription of endogenous p21 as well as muscle-specific genes. Specifically, we present evidence that HDAC1 can bind directly to MyoD and use an acetylated MyoD as a substrate in vitro, whereas a mutant version of HDAC1 (H141A) can not. Further more, this mutant also fails to repress MyoD-mediated transcription in vivo, and unlike wild-type HDAC1 it can not inhibit myogenic conversion, as judged by confocal microscopy. Finally, we show that an endogenous MyoD can be acetylated upon its conversion to a hypophosphorylated state and only when the cells have been induced to differentiate. These results provide for a model which postulates that MyoD may be co-dependent on HDAC1 and P/CAF for temporally controlling its transcriptional activity before and after the differentiation of muscle cells.


Subject(s)
Histone Deacetylases/physiology , Muscle, Skeletal/physiology , MyoD Protein/metabolism , Transcription, Genetic , Acetylation , Acetyltransferases/genetics , Acetyltransferases/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Differentiation , Cell Line , Cell Line, Transformed , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , Histone Acetyltransferases , Histone Deacetylase 1 , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Jurkat Cells , Mice , Muscle, Skeletal/cytology , MyoD Protein/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors , p300-CBP Transcription Factors
3.
J Cancer Res Clin Oncol ; 126(4): 185-90, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10782890

ABSTRACT

PURPOSE: Genomic stability is maintained by error-free DNA replication, repair, and recombination. To determine if repair genes contribute to genomic instability, we used a newly established cell line RCC-AJR (from clear-cell renal cell carcinoma) to examine hMSH2 (a mismatch-repair gene) and the gene encoding DNA beta polymerase (polbeta; a known contributor to base-excision repair). METHODS: Coding sequences of hMSH2 and polbeta were amplified by the polymerase chain reaction (PCR) using RNA from RCC-AJR cells and matched normal kidney (NK) cells from the same patient. Nucleotide sequences of the PCR products were determined by the dideoxy-DNA method and direct sequencing. Expressions of repair genes were assayed by Western blotting. Microsatellite stability in RCC-AJR cells was assayed by alteration in (CA)n repeats. RESULTS: In the RCC-AJR cells, we detected (a) a deletion of 1476 bp encoding 492 amino acids of hMSH2 cDNA, (b) an 87-bp deletion in the polbeta coding sequence, (c) truncated forms of hMSH2 and polbeta proteins, and (d) microsatellite instability. CONCLUSIONS: This study provides evidence of alterations in hMSH2 and polbeta in the homogeneous cell population of an RCC-AJR tumor culture. The data indicate that repair genes may help preserve genomic stability in this cell line. We believe that this new primary RCC-AJR cell line will prove a useful model for investigating the cascade of genetic events in renal cells that leads to renal carcinogenesis.


Subject(s)
Carcinoma, Renal Cell/genetics , DNA Polymerase beta/genetics , DNA Repair/genetics , DNA-Binding Proteins , Kidney Neoplasms/genetics , Proto-Oncogene Proteins/genetics , Carcinoma, Renal Cell/pathology , Gene Deletion , Humans , Kidney Neoplasms/pathology , Microsatellite Repeats/genetics , MutS Homolog 2 Protein , Polymerase Chain Reaction , Tumor Cells, Cultured
4.
J Cell Biol ; 149(2): 281-92, 2000 Apr 17.
Article in English | MEDLINE | ID: mdl-10769022

ABSTRACT

During differentiation, skeletal muscle cells withdraw from the cell cycle and fuse into multinucleated myotubes. Unlike quiescent cells, however, these cells cannot be induced to reenter S phase by means of growth factor stimulation. The studies reported here document that both the retinoblastoma protein (Rb) and the cyclin-dependent kinase (cdk) inhibitor p21 contribute to this unresponsiveness. We show that the inactivation of Rb and p21 through the binding of the adenovirus E1A protein leads to the induction of DNA replication in differentiated muscle cells. Moreover, inactivation of p21 by E1A results in the restoration of cyclin E-cdk2 activity, a kinase made nonfunctional by the binding of p21 and whose protein levels in differentiated muscle cells is relatively low in amount. We also show that restoration of kinase activity leads to the phosphorylation of Rb but that this in itself is not sufficient for allowing differentiated muscle cells to reenter the cell cycle. All the results obtained are consistent with the fact that Rb is functioning downstream of p21 and that the activities of these two proteins may be linked in sustaining the postmitotic state.


Subject(s)
Adenovirus E1A Proteins/metabolism , CDC2-CDC28 Kinases , Cell Differentiation/physiology , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , DNA Replication/physiology , Protein Serine-Threonine Kinases/metabolism , Retinoblastoma Protein/metabolism , Animals , Cell Line , Cell-Free System , Cyclin E/metabolism , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinases/antagonists & inhibitors , Mice , Muscle, Skeletal/cytology , Muscle, Skeletal/physiology , Mutagenesis , Recombinant Proteins/metabolism , Transfection
5.
J Clin Invest ; 101(1): 153-9, 1998 Jan 01.
Article in English | MEDLINE | ID: mdl-9421477

ABSTRACT

Small cell lung cancer (SCLC) accounts for 25% of all lung cancers, and is almost uniformly fatal. Unlike other lung cancers, ras mutations have not been reported in SCLC, suggesting that activation of ras-associated signal transduction pathways such as the raf-MEK mitogen-activated protein kinases (MAPK) are associated with biological consequences that are unique from other cancers. The biological effects of raf activation in small cell lung cancer cells was determined by transfecting NCI-H209 or NCI-H510 SCLC cells with a gene encoding a fusion protein consisting of an oncogenic form of human Raf-1 and the hormone binding domain of the estrogen receptor (DeltaRaf-1:ER), which can be activated with estradiol. DeltaRaf-1:ER activation resulted in phosphorylation of MAPK. Activation of this pathway caused a dramatic loss of soft agar cloning ability, suppression of growth capacity, associated with cell accumulation in G1 and G2, and S phase depletion. Raf activation in these SCLC cells was accompanied by a marked induction of the cyclin-dependent kinase (cdk) inhibitor p27(kip1), and a decrease in cdk2 protein kinase activities. Each of these events can be inhibited by pretreatment with the MEK inhibitor PD098059. These data demonstrate that MAPK activation by DeltaRaf-1:ER can activate growth inhibitory pathways leading to cell cycle arrest. These data suggest that raf/MEK/ MAPK pathway activation, rather than inhibition, may be a therapeutic target in SCLC and other neuroendocrine tumors.


Subject(s)
CDC2-CDC28 Kinases , Carcinoma, Small Cell/pathology , Cell Cycle Proteins , Lung Neoplasms/pathology , Proto-Oncogene Proteins c-raf/metabolism , Tumor Suppressor Proteins , Adenovirus E1A Proteins/pharmacology , CDC2 Protein Kinase/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Cycle , Cell Division , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , Enzyme Inhibitors/metabolism , Estradiol/pharmacology , Flavonoids/pharmacology , Humans , Microtubule-Associated Proteins/metabolism , Mitogen-Activated Protein Kinase Kinases , Phosphorylation , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-raf/biosynthesis , Proto-Oncogene Proteins c-raf/genetics , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology , Tumor Cells, Cultured
6.
J Virol ; 70(5): 2911-21, 1996 May.
Article in English | MEDLINE | ID: mdl-8627766

ABSTRACT

The adenovirus E1A protein of 243 amino acids has been shown to affect a variety of cellular functions, most notably the immortalization of primary cells and the promotion of quiescent cells into S phase. The activity of E1A is derived, in part, from its association with various cellular proteins, many of which play important roles in regulating cell cycle progression. E1A is known to have multiple sites of phosphorylation. It has been suggested that cell cycle-dependent phosphorylation may also control some of E1A's functions. We find now that immune complexes of cyclin-dependent kinases such as cdk4, cdk2, and cdc2 are all capable of phosphorylating E1A in vitro. Additionally, the sites on E1A phosphorylated by these kinases in vitro are similar to the E1A sites phosphorylated in vivo. We have also found that a phosphorylated E1A is far more efficient than an unphosphorylated E1A in associating with pRB and in disrupting E2F/DP-pRB complexes as well. On the basis of our findings and the differences in timing and expression levels of the various cyclins regulating cdks, we suggest that E1A functions at different control points in the cell cycle and that phosphorylation controls, to some extent, its biological functions.


Subject(s)
Adenovirus E1A Proteins/metabolism , Carrier Proteins , Cell Cycle Proteins , Cyclin-Dependent Kinases/metabolism , DNA-Binding Proteins , Retinoblastoma Protein/metabolism , Transcription Factors/metabolism , Adenovirus E1A Proteins/isolation & purification , Base Sequence , Binding Sites , Cell Nucleus/metabolism , E2F Transcription Factors , Glutathione Transferase/biosynthesis , HeLa Cells , Humans , Kinetics , Macromolecular Substances , Molecular Sequence Data , Oligonucleotide Probes , Phosphoproteins/isolation & purification , Phosphoproteins/metabolism , Phosphorylation , Protein Binding , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Retinoblastoma Protein/biosynthesis , Retinoblastoma Protein/isolation & purification , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1 , Transcription Factors/antagonists & inhibitors , Transcription Factors/isolation & purification
7.
J Biol Chem ; 271(16): 9376-83, 1996 Apr 19.
Article in English | MEDLINE | ID: mdl-8621603

ABSTRACT

Prostaglandin A2 (PGA2) reversibly blocked the cell cycle progression of NIH 3T3 cells at G1 and G2/M phase. When it was applied to cells synchronized in G0 or S phase, cells were blocked at G1 and G2/M, respectively. The G2/M blockage was transient. Microinjected oncogenic leucine 61 Ras protein could not override the PGA2 induced G1 blockage, nor could previous transformation with the v-raf oncogene. The serum-induced activation of mitogen-activated protein kinase was not inhibited by PGA2 treatment. These data suggest that PGA2 blocks cell cycle progression without interfering with the cytosolic proliferative signaling pathway. Combined microinjection of E2F-1 and DP-1 proteins or microinjected adenovirus E1A protein, however, could induce S phase in cells arrested in G1 by PGA2, indicating that PGA2 does not directly inhibit the process of DNA synthesis. In quiescent cells, PGA2 blocked the normal hyperphosphorylation of the retinoblastoma susceptible gene product and the activation of cyclin-dependent kinase (CDK) 2 and CDK4, in response to serum stimulation. PGA2 treatment elevated the p21Waf1/Cip1/Sdi1 protein expression level. These data indicate that PGA2 may arrest the cell cycle in G1 by interfering with the activation of G1 phase CDKs.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Cycle , Cyclin-Dependent Kinases/metabolism , Prostaglandins A/pharmacology , 3T3 Cells , Animals , Cell Cycle/drug effects , Cell Transformation, Neoplastic , Cyclin-Dependent Kinases/antagonists & inhibitors , Enzyme Activation , G1 Phase , G2 Phase , Kinetics , Mice , Microinjections , Mitosis , Oncogene Proteins v-raf , Oncogenes , Phosphorylation , Retinoblastoma Protein/isolation & purification , Retinoblastoma Protein/metabolism , Retroviridae Proteins, Oncogenic/biosynthesis , Retroviridae Proteins, Oncogenic/genetics , Time Factors , ras Proteins/biosynthesis , ras Proteins/metabolism
8.
Nature ; 380(6571): 262-5, 1996 Mar 21.
Article in English | MEDLINE | ID: mdl-8637577

ABSTRACT

The adenovirus oncoprotein E1A and the simian virus SV40 large T antigen can both reverse the strong growth-inhibitory effect of transforming growth factor(TGF)-beta on mink lung epithelial cells: exposure of TGF-beta causes these cells to arrest late in the G1 phase of the cell cycle (ref. 3). This arrest correlates with an increase in expression of the protein p15Ink4B (ref. 4), inactivation of the cyclin E/A-cdk2 complex by the inhibitory protein p27Kip1 (refs 5-7), and with the accumulation of unphosphorylated retinoblastoma protein. The rescue by E1A of cells from TGF-beta arrest is partly independent of its binding to retinoblastoma protein. Here we show that E1A directly affects the cyclin-dependent kinase inhibitor p27Kip1 in TGF-beta-treated cells by binding to it and blocking its inhibitory effect, thereby restoring the activity of the cyclin-cdk2 kinase complex. In this way, E1A can overcome the effect of TGF-beta and modulate the cell cycle. To our knowledge, E1A provides the first example of a viral oncoprotein that can disable a cellular protein whose function is to inhibit the activity of cyclin-dependent kinases.


Subject(s)
Adenovirus E1A Proteins/pharmacology , CDC2-CDC28 Kinases , Cell Cycle Proteins , Enzyme Inhibitors , Microtubule-Associated Proteins/antagonists & inhibitors , Transforming Growth Factor beta/pharmacology , Tumor Suppressor Proteins , Animals , Cell Line , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Mink , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism
9.
Nature ; 374(6517): 85-8, 1995 Mar 02.
Article in English | MEDLINE | ID: mdl-7870179

ABSTRACT

The 265K nuclear protein CBP was initially identified as a co-activator for the protein kinase A (PKA)-phosphorylated form of the transcription factor CREB. The domains in CBP that are involved in CREB binding and transcriptional activation are highly related to the adenoviral E1A-associated cellular protein p300 (refs 2, 3), and to two hypothetical proteins from Caenorhabditis elegans, R10E11.1 and K03H1.10 (refs 4 and 5, respectively), whose functions are unknown. Here, we show that CBP and p300 have similar binding affinity for the PKA-phosphorylated form of CREB, and that p300 can substitute for CBP in potentiating CREB-activated gene expression. We find that E1A binds to CBP through a domain conserved with p300 and represses the CREB-dependent co-activator functions of both CBP and p300. Our results indicate that the gene repression and cell immortalization functions associated with E1A involve the inactivation of a family of related proteins that normally participate in second-messenger-regulated gene expression.


Subject(s)
Adenovirus E1A Proteins/metabolism , Nuclear Proteins/metabolism , Trans-Activators , Transcription Factors/metabolism , Transcriptional Activation , Amino Acid Sequence , CREB-Binding Protein , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , E1A-Associated p300 Protein , Molecular Sequence Data , Phosphorylation , Protein Binding , Sequence Homology, Amino Acid , Tumor Cells, Cultured
10.
EMBO J ; 13(24): 6107-14, 1994 Dec 15.
Article in English | MEDLINE | ID: mdl-7813447

ABSTRACT

The adenovirus E1A protein can induce cellular DNA synthesis in growth-arrested cells by interacting with the cellular protein p300 or pRb. In addition, serum- and growth factor-dependent cells require ras activity to initiate DNA synthesis and recently we have shown that Balb/c 3T3 cells can be blocked in either early or late G1 following microinjection of an anti-ras antibody. In this study, the E1A 243 amino acid protein is shown through microinjection not only to shorten the G0 to S phase interval but, what is more important, to override the inhibitory effects exerted by the anti-ras antibody in either early or late G1. Specifically, whether E1A is co-injected with anti-ras into quiescent cells or injected 18 h following a separate injection of anti-ras after serum stimulation, it efficiently induces cellular DNA synthesis in cells that would otherwise be blocked in G0/G1. Moreover, injection of a mutant form of E1A that can no longer associate with p300 is just as efficient as wild-type E1A in stimulating DNA synthesis in cells whose ras activity has been neutralized by anti-ras. The results presented here show that E1A is capable of overriding the requirement of cellular ras activity in promoting the entry of cells into S phase. Moreover, the results suggest the possibility that pRb and/or pRb-related proteins may function in a ras-dependent pathway that enables E1A to achieve this activity.


Subject(s)
Adenovirus E1A Proteins/metabolism , Cell Cycle/physiology , DNA/biosynthesis , Proto-Oncogene Proteins p21(ras)/metabolism , Trans-Activators , 3T3 Cells , Adenovirus E1A Proteins/immunology , Animals , Antibodies, Monoclonal/pharmacology , Cell Cycle/drug effects , E1A-Associated p300 Protein , G1 Phase/physiology , Mice , Mice, Inbred BALB C , Microinjections , Nuclear Proteins/metabolism , Resting Phase, Cell Cycle/physiology , Retinoblastoma Protein/metabolism , S Phase/physiology , Transcription Factors/metabolism
11.
Oncogene ; 9(9): 2605-12, 1994 Sep.
Article in English | MEDLINE | ID: mdl-8058324

ABSTRACT

E2F is a cellular transcription factor that is regulated during the cell cycle through interactions with the product of the retinoblastoma susceptibility gene (RB1) and the pRb-like p107 and p130 proteins. Analysis of mutations within both adenovirus E1A and pRb, which affected their ability to regulate cellular proliferation and alter E2F activity, suggested that E2F may play a role in cell cycle progression. Microinjection of a GST-E2F-1 fusion protein into quiescent Balb/c 3T3 cells induced DNA synthesis whereas co-injection of GST-E2F-1 and GST-E2F(95-191) protein, encoding only the DNA binding domain of E2F-1, blocked the induction of S-phase. While E1A likely targets multiple cellular pathways, co-injection of the GST-E2F(95-191) dominant inhibitory protein with 12S E1A protein blocked E1A-mediated induction of DNA synthesis, suggesting that the E2F-dependent pathway is dominant. Analysis of the interval required for microinjected quiescent cells to enter S-phase indicated that E2F-1 acted faster than either E1A or serum.


Subject(s)
Adenovirus E1A Proteins/physiology , Carrier Proteins , Cell Cycle Proteins , Cell Cycle , DNA-Binding Proteins , Transcription Factors/physiology , 3T3 Cells , Animals , DNA/biosynthesis , E2F Transcription Factors , E2F1 Transcription Factor , Genes, myc , Glutathione Transferase/genetics , HeLa Cells , Humans , Mice , Mutation , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1 , Transcription Factors/genetics
12.
Oncogene ; 9(6): 1733-7, 1994 Jun.
Article in English | MEDLINE | ID: mdl-8183570

ABSTRACT

A protein of 300 kDa (p300) associates with the adenovirus E1A proteins and has been implicated in the control of cell cycle progression. In mammalian cells, p300 is actively phosphorylated in both quiescent and proliferating cells and its level of phosphorylation increases as it travels from late G1 into M phase. E1A requires p300 for the induction of cellular DNA synthesis and the repression of enhancer mediated transcription, suggesting that p300 may be involved in pathways that are important to cell proliferation and gene expression. Since the activities of most cell cycle regulatory proteins depend on their phosphorylation state, the possibility exists that certain activities of p300 might also be controlled by phosphorylation and that E1A might in fact be affecting these events. We show here by in vitro analysis that E1A inhibits the phosphorylation of p300 by decreasing the rate of incorporation of phosphate into p300. We also show that p300 can be used as a substrate for the cyclin-dependent p33cdk2 and p34cdc2 kinases, and propose that E1A might be antagonistic to these enzymes in phosphorylating p300. Thus, these results indicate a possible novel function by which E1A can interfere with cellular pathways.


Subject(s)
Adenovirus E1A Proteins/pharmacology , Nuclear Proteins/metabolism , Adenosine Triphosphate/metabolism , DNA/biosynthesis , HeLa Cells , Humans , Phosphorylation
13.
Protein Expr Purif ; 3(3): 246-55, 1992 Jun.
Article in English | MEDLINE | ID: mdl-1392621

ABSTRACT

The E1B 175-amino-acid (175R) protein of adenovirus 2 is required for cellular transformation of primary cells and establishing cell morphology in lytically infected cells. To investigate the biochemical function of this protein, we constructed a bacterial expression vector (pKHB1-T) to produce the 175R protein in sufficient amounts for purification and biochemical analysis. On the basis of DNA sequencing, gel electrophoresis, and immunoblot analysis, the pKHB1-T-encoded 175R protein appears to be identical to that expressed transiently in mammalian or adenovirus-transformed cells. The bacterially produced viral protein was also found to be quite stable and without any modifications. Partial purification of the pKHB1-T-encoded protein revealed that the majority of its associates with the inner membrane of the bacterial cell. This, together with the possibility of the 175R protein containing an N-terminal amphipathic alpha-helix as a potential translocation signal, suggests that there may be a common mechanism of protein transport operating in both eucaryotic and procaryotic systems.


Subject(s)
Adenoviridae/genetics , Adenovirus E1B Proteins/biosynthesis , Escherichia coli/chemistry , Membrane Proteins/biosynthesis , Recombinant Fusion Proteins/biosynthesis , Adenovirus E1B Proteins/isolation & purification , Amino Acid Sequence , Animals , Base Sequence , Cells, Cultured , Chlorocebus aethiops , Fibroblasts , Gene Expression , Genetic Vectors , Membrane Proteins/isolation & purification , Molecular Sequence Data , Promoter Regions, Genetic , Recombinant Fusion Proteins/isolation & purification
14.
Protein Expr Purif ; 3(1): 8-17, 1992 Feb.
Article in English | MEDLINE | ID: mdl-1422211

ABSTRACT

The adenovirus E1A 243R protein immortalizes primary cells in culture and induces part of the phenotypes required for transformation. It has also been shown to interact with a number of cellular polypeptides, including the product of the retinoblastoma gene. To understand more fully the molecular activities of the E1A 243R protein in association with these proteins as well as its role in the processes of cellular growth, we have developed a method for rapidly purifying this protein from genetically engineered Escherichia coli under nondenaturing conditions. The plasmid-encoded E1A protein, when expressed in a protease-deficient mutant, is found to have the same length and amino acid sequence as that which is produced in a mammalian cell. The procedure for purifying the E1A 243R protein from bacteria relies primarily upon immunoaffinity chromatography and the use of a peptide comprising the epitope recognized by an E1A-specific antibody. Elution of the E1A protein under this condition allows for gentle isolation and a purity that ranges from 90 to 96%. However, without the addition of micromolar amounts of ATP prior to its elution from the antibody column, the E1A protein is found in association with an E. coli protein of 70 kDa. Immunoblot analysis with a specific antibody showed that this bacterial protein was the heat shock protein dnaK, which is known to have extensive homology with the hsp-hsc70 family of proteins in mammalian cells. Recognition of E1A by the dnaK protein may very well reflect a situation that also occurs between the mammalian heat shock proteins and the E1A 243R protein after adenovirus infection.


Subject(s)
Adenovirus E1A Proteins/isolation & purification , Bacterial Proteins/isolation & purification , Escherichia coli Proteins , HSP70 Heat-Shock Proteins , Heat-Shock Proteins/isolation & purification , Amino Acid Sequence , Escherichia coli/chemistry , Heat-Shock Proteins/genetics , Molecular Sequence Data , Recombinant Proteins/genetics
15.
J Biol Chem ; 267(4): 2530-6, 1992 Feb 05.
Article in English | MEDLINE | ID: mdl-1310318

ABSTRACT

Adenovirus infection of hepatoma cells inhibited transcription of the phosphoenolpyruvate carboxykinase (GTP) (EC 4.1.1.32) (PEPCK) gene and virtually eliminated transcription of a chimeric gene which contained the PEPCK promoter linked to the structural gene for chloramphenicol acetyltransferase (CAT). This effect is due to the viral protein E1A, since adenovirus containing a deletion in the E1A gene did not repress transcription from the PEPCK promoter. Both the 243R and 283R products of the E1A gene were effective. The conserved region 1 (CR-1) domain of E1A was required for this effect. Treatment of hepatoma cells with 8-bromo-cAMP or transfection with plasmids coding for the catalytic subunit of protein kinase A, CAAT/enhancer binding protein alpha (C/EBP), or Jun, all potent inducers of PEPCK gene transcription, did not relieve the inhibition caused by E1A. This inhibition does not appear to be mediated by major enhancer elements and in the PEPCK gene since transcription from the PEPCK promoter containing block mutations in binding domains for C/EBP and cAMP regulatory element binding protein (CREB) was also inhibited by E1A. Transcription of chimeric genes containing two copies each of the major cAMP response domains (CRE-1 and P-3) linked to a neutral promoter and fused to the CAT structural gene was stimulated by the catalytic subunit of protein kinase A, but this effect was totally inhibited by E1A. The strong repressive effect of E1A on PEPCK gene transcription seems to involve an interruption of an obligatory interaction between factors which bind to the cAMP response element in the PEPCK promoter and the TATA box.


Subject(s)
Oncogene Proteins, Viral/metabolism , Phosphoenolpyruvate Carboxykinase (GTP)/genetics , Transcription, Genetic , Adenoviridae/isolation & purification , Adenovirus Early Proteins , CCAAT-Enhancer-Binding Proteins , Carcinoma, Hepatocellular , Chloramphenicol O-Acetyltransferase/genetics , Chloramphenicol O-Acetyltransferase/metabolism , Cyclic AMP/pharmacology , Cyclic AMP Response Element-Binding Protein , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Viral , Mutation , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Protein Kinases/metabolism , Proto-Oncogene Proteins c-jun/metabolism , TATA Box , Transcription Factors/metabolism , Tumor Cells, Cultured
16.
J Virol ; 65(10): 5297-304, 1991 Oct.
Article in English | MEDLINE | ID: mdl-1832723

ABSTRACT

We have previously demonstrated that a purified bacterially synthesized E1A 289-amino-acid protein is capable of stimulating transcription from the promoters of genes transcribed by RNA polymerase II in vitro (R. Spangler, M. Bruner, B. Dalie, and M. L. Harter, Science 237:1044-1046, 1987). In this study, we show that this protein is also capable of transactivating in vitro the adenovirus virus-associated (VA1) RNA gene transcribed by RNA polymerase III. Pertinent to the transcription of this gene is the rate-limiting component, TFIIIC, which appears to be of two distinct forms in uninfected HeLa cells. The addition of an oligonucleotide containing a TFIIIC binding site to HeLa whole-cell extracts inhibits VA1 transcription by sequestering TFIIIC. However, the addition of purified E1A to extracts previously challenged with the TFIIIC oligonucleotide restores the level of VA1 transcription. When included in the same reaction, an E1A-specific monoclonal antibody reverses the restoration. Incubation of purified E1A with either HeLa cell nuclear or whole-cell extracts alters the DNA-binding properties of TFIIIC as detected by gel shift assays. This alteration does not occur if E1A-specific antibody and E1A protein are added simultaneously to the extract. In contrast, the addition of this antibody to extracts at a later time does not reverse the alteration observed in the TFIIIC binding activities. Never at any time did we note the formation of novel TFIIIC-promoter complexes after the addition of E1A to nuclear extracts. These results clearly establish that E1A mediates its effect on VA1 transcription through TFIIIC in a very rapid yet indirect manner.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Adenoviruses, Human/genetics , Oncogene Proteins, Viral/metabolism , RNA Polymerase III/metabolism , Transcription Factors, TFIII , Transcription Factors/metabolism , Transcription, Genetic , Adenovirus Early Proteins , Adenoviruses, Human/metabolism , Base Sequence , Gene Expression Regulation, Viral , HeLa Cells/physiology , Humans , Molecular Sequence Data , Oligonucleotide Probes , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/isolation & purification , Plasmids , Promoter Regions, Genetic , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Transcription Factors/genetics
17.
Proc Natl Acad Sci U S A ; 88(17): 7459-63, 1991 Sep 01.
Article in English | MEDLINE | ID: mdl-1652751

ABSTRACT

Infection with wild-type adenovirus 5, but not with a mutant lacking the E1A gene, prevented the induction by interferon (IFN) alpha of chloramphenicol acetyltransferase (CAT) activity in HeLaM cell lines that had been permanently transfected with chimeric CAT reporter genes driven by the transcriptional regulatory regions of the IFN-responsive genes 561 and 6-16. Similar inhibition of IFN-inducible CAT activity was observed in cells that were cotransfected with the same reporter genes and plasmids expressing either the E1A 289- or 243-amino acid protein. These proteins also prevented the induction of CAT activity by IFN-gamma from a cotransfected HLA-DR alpha-CAT gene. Experiments with E1A mutants mapped the inhibitory activity to amino acid residues 38-65 of these proteins. In a HeLa cell line permanently expressing the E1A 289-amino acid protein, the replication of vesicular stomatitis virus and encephalomyocarditis virus was not inhibited by IFN-alpha, suggesting a global blockade of IFN responses. In accord with this theory, induction of 561, 1-8, and (2'-5')oligoadenylate synthetase mRNAs by IFN was blocked in these cells at the transcriptional level. The observed transcriptional inhibition could be attributed to the lack of formation of the crucial IFN-stimulated gene factor 3 (ISGF3) transcriptional complex. As shown by mobility shift assays, this complex was not formed in the nuclear extracts of IFN-treated adenovirus-infected cells or IFN-treated E1A-producing cells. These nuclear extracts were deficient in both ISGF3 alpha and ISGF3 gamma subunits. However, they did not block the formation of ISGF3 complex from exogenously added components.


Subject(s)
Adenoviruses, Human/genetics , Interferon Type I/pharmacology , Oncogene Proteins, Viral/metabolism , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Vesicular stomatitis Indiana virus/physiology , Adenovirus Early Proteins , Adenoviruses, Human/metabolism , Blotting, Northern , Cell Transformation, Viral , Chimera , Chloramphenicol O-Acetyltransferase/genetics , Chloramphenicol O-Acetyltransferase/metabolism , Gene Expression/drug effects , Genetic Vectors , HeLa Cells/physiology , Humans , Kinetics , Oncogene Proteins, Viral/genetics , Plasmids , Transfection , Vesicular stomatitis Indiana virus/drug effects , Vesicular stomatitis Indiana virus/genetics , Virus Replication/drug effects
18.
EMBO J ; 7(3): 835-41, 1988 Mar.
Article in English | MEDLINE | ID: mdl-2969333

ABSTRACT

An adenovirus 2 289 amino acid (289R) E1A protein purified from Escherichia coli has been shown to interact with DNA by two independent methods. UV-crosslinking of complexes containing unmodified, uniformly 32P-labelled DNA and purified E1A protein induced efficient labelling of the protein with covalently attached oligonucleotides, indicating that the E1A protein itself contacts DNA. Discrete nucleoprotein species were also observed when E1A protein--DNA complexes were analysed by gel electrophoresis. Although the 289R E1A protein exhibited no significant binding to single-stranded DNA or to RNA, no evidence for its sequence-specific binding to double-stranded DNA was obtained with either assay. Identification of the sites of covalent attachment of 32P-labelled oligonucleotides by partial proteolysis of the crosslinked E1A protein indicated that the interaction of this protein with DNA is mediated via domain(s) in the C-terminal half of the protein. Such previously unrecognized DNA-binding activity is likely to contribute to the regulatory activities of this important adenoviral protein.


Subject(s)
DNA, Viral/metabolism , DNA-Binding Proteins/metabolism , Oncogene Proteins, Viral/metabolism , Adenovirus Early Proteins , Binding Sites
19.
J Biol Chem ; 263(8): 3984-9, 1988 Mar 15.
Article in English | MEDLINE | ID: mdl-2964445

ABSTRACT

The adenovirus 2 E1A gene encodes a multifunctional protein of 289 amino acids that can immortalize primary rodent cells and transcriptionally activate a number of viral and cellular genes. To facilitate an understanding of the molecular basis for the various actions of E1A, we have redesigned our bacterial expression vector (Ko, J.-L., and Harter, M. L. (1984) Mol. Cell. Biol. 4, 1427-1439) containing the cloned E1A gene such that a soluble authentic E1A protein now constitutes approximately 1.5% of the total Escherichia coli cellular protein. Further, we have developed a simple rapid purification scheme without the use of detergents or denaturants and show a purity of greater than 98% with a yield of approximately 53%. The E1A so purified is biologically active, stimulating cellular DNA synthesis following microinjection into quiescent NIH 3T3 and REF52 cells. In another report (Spangler, R., Bruner, M., Dalie, B., and Harter, M. L. (1987) Science 237, 1044-1046) we have also shown that our purified E1A protein activates transcription from appropriate promoters in an in vitro system.


Subject(s)
Adenoviruses, Human/genetics , Antigens, Viral/genetics , Escherichia coli/genetics , Oncogene Proteins, Viral/genetics , Adenovirus Early Proteins , Adenoviruses, Human/immunology , Animals , Cell Line , Cells, Cultured , DNA Replication , Genes , Genes, Viral , Mice , Oncogene Proteins, Viral/isolation & purification , Plasmids , Promoter Regions, Genetic
20.
Science ; 237(4818): 1044-6, 1987 Aug 28.
Article in English | MEDLINE | ID: mdl-2956686

ABSTRACT

The primary product of the adenovirus E1A gene is a protein that is sufficient for controlling host-cell proliferation and immortalizing primary rodent cells. The mechanism by which the protein induces these cellular effects is poorly understood, but might be linked to its ability to regulate RNA transcription from a number of viral and cellular genes. The mechanism of E1A's transcriptional-activation (trans-activation) was studied here by monitoring the protein's effect on specific adenovirus promoters in two types of transcriptional systems in vitro. One of these systems consisted of extracts from transformed cells constitutively expressing E1A, and the other consisted of extracts of HeLa cells supplemented with a plasmid-encoded E1A protein purified from Escherichia coli. The results show that the E1A protein specifically stimulates transcription from adenovirus promoters; thus, the induction of cellular transcription factors is not necessary to explain the stimulation of transcription by E1A.


Subject(s)
Adenoviridae/genetics , Oncogene Proteins, Viral/pharmacology , Promoter Regions, Genetic/drug effects , Adenovirus Early Proteins , Cell-Free System , Escherichia coli , HeLa Cells , Humans , Virus Activation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...