Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
1.
Mol Cell Biol ; 6(7): 2392-401, 1986 Jul.
Article in English | MEDLINE | ID: mdl-3785199

ABSTRACT

We have developed an in vitro transcription system for the murine dihydrofolate reductase gene. Although transcription in vitro from a linearized template was initiated at the same start sites as in vivo, the correct ratios were more closely approximated when a supercoiled template was used. In addition, whereas the dihydrofolate reductase promoter functions bidirectionally in vivo, the initiation signals directed unidirectional transcription in this in vitro system. The dihydrofolate reductase gene does not have a typical TATA box, but has four GGGCGG hexanucleotides within 300 base pairs 5' of the AUG codon. Deletion analysis suggested that, although sequences surrounding each of the GC boxes could specify initiation approximately 40 to 50 nucleotides downstream, three of the four GC boxes could be removed without changing the accuracy or efficiency of initiation at the major in vivo site. The dihydrofolate reductase promoter initiated transcription very rapidly in vitro, with transcripts visible by 1 min and almost maximal by 2 min at 30 degrees C with no preincubation. Nuclear extracts prepared from cells blocked in the S phase by aphidicolin or from adenovirus-infected cells at 16 h postinfection had enhanced dihydrofolate reductase transcriptional activity. This increased in vitro transcription mimicked the increase in dihydrofolate reductase mRNA seen in S-phase cells and suggested the presence of a cell-cycle-specific factor(s) which stimulated transcription from the dihydrofolate reductase gene.


Subject(s)
Promoter Regions, Genetic , Tetrahydrofolate Dehydrogenase/genetics , Transcription, Genetic , Adenoviridae , Animals , Base Sequence , Chromosome Deletion , Electrophoresis, Polyacrylamide Gel , HeLa Cells , Humans , Magnesium/pharmacology , Magnesium Chloride , Mice , Nucleic Acid Conformation , Temperature , Time Factors
2.
Mol Cell Biol ; 6(2): 365-71, 1986 Feb.
Article in English | MEDLINE | ID: mdl-3785152

ABSTRACT

The murine dihydrofolate reductase gene codes for mRNAs that differ in the length of their 3' untranslated region as well as in the length of their 5' leader sequence. In addition, the dihydrofolate reductase promoter functions bidirectionally, producing a series of RNAs from the opposite strand than the dihydrofolate reductase mRNAs. We have examined the production of these RNAs and their heterogeneous 5' and 3' termini as mouse 3T6 cells progress through a physiologically continuous cell cycle. We found that all of the transcripts traverse the cell cycle in a similar manner, increasing at the G1/S boundary without significantly changing their ratios relative to one another. We conclude that cell-cycle regulation of dihydrofolate reductase is achieved without recruiting new transcription initiation sites and without a change in polyadenylation sites. It appears that the mechanism responsible for the transcriptional cell-cycle regulation of the dihydrofolate reductase gene is manifested only by transiently increasing the efficiency of transcription at the dihydrofolate reductase promoter.


Subject(s)
Genes , RNA, Messenger/genetics , Tetrahydrofolate Dehydrogenase/genetics , Transcription, Genetic , Animals , Cell Cycle , Cell Line , Drug Resistance , Methotrexate/pharmacology , Mice , Plasmids
3.
Mol Cell Biol ; 17(5): 2529-37, 1997 May.
Article in English | MEDLINE | ID: mdl-9111322

ABSTRACT

Carbamoyl-phosphate synthase/aspartate carbamoyltransferase/dihydroorotase, which is encoded by the cad gene, is required for the first three rate-limiting steps of de novo pyrimidine biosynthesis. It has been previously demonstrated that cad transcription increases at the G1/S-phase boundary, as quiescent cells reenter the proliferative cell cycle. The growth-responsive element has been mapped to an E box at +65 in the hamster cad promoter. Using an in vivo UV cross-linking and immunoprecipitation assay, we show that Myc, Max, and upstream stimulatory factor (USF) bind to the chromosomal cad promoter. To determine whether binding of Myc-Max or USF is critical for cad growth regulation, we analyzed promoter constructs which contain mutations in the nucleotides flanking the E box. We demonstrate that altering nucleotides which flank the cad E box to sequences which decrease Myc-Max binding in vitro correlates with a loss of cad G1/S-phase transcriptional activation. This result supports the conclusion that binding of Myc-Max, but not USF, is essential for cad regulation. Our investigations demonstrate that the endogenous cad E box can be bound by more than one transcription factor, but growth-induced cad expression is achieved only by Myc.


Subject(s)
Aspartate Carbamoyltransferase/genetics , Carbamoyl-Phosphate Synthase (Glutamine-Hydrolyzing)/genetics , DNA-Binding Proteins/genetics , Dihydroorotase/genetics , Genes, myc/genetics , Helix-Loop-Helix Motifs , Multienzyme Complexes/genetics , Neoplasm Proteins/genetics , Promoter Regions, Genetic , Transcription Factors/genetics , 3T3 Cells , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Basic-Leucine Zipper Transcription Factors , Binding Sites , Consensus Sequence , Cricetinae , DNA/metabolism , G1 Phase , Mice , Models, Molecular , S Phase , Ultraviolet Rays , Upstream Stimulatory Factors
4.
Mol Cell Biol ; 19(12): 8393-9, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10567564

ABSTRACT

Previously, we have used a chromatin cross-linking and immunoprecipitation protocol for the analysis of Myc and USF binding to the cad promoter. The adaptation of this technique for the study of mammalian transcription factors was a big step forward in the analysis of transcription factor family member specificity, allowing for the first time a definitive knowledge of which factor binds to a promoter region under normal physiological conditions. However, due to limitations of the assay, our previous studies could not definitively prove that both Myc and USF bound to the exact same site on the cad promoter, nor could we directly correlate loss of in vivo binding of a particular factor with loss of transcriptional activity. Therefore, we have further modified the chromatin immunoprecipitation protocol to alleviate these problems. We have now shown that it is possible to coexamine growth-regulated transcriptional activity and promoter occupancy by using stably integrated promoter constructs. We show that both Myc and USF bind to the exact same E box on the cad promoter, suggesting that competition between these two factors for a single site occurs in living cells. We also find that cad promoter constructs that retain USF binding but lose Myc binding in vivo no longer display an increase in transcriptional activity in mid- to late G(1) phase of the cell cycle. Finally, we propose that cell cycle-regulated transcriptional activation of the cad promoter may be a stochastic, rather than a predetermined, process.


Subject(s)
DNA-Binding Proteins , Neoplasm Proteins/metabolism , Phosphopyruvate Hydratase , Promoter Regions, Genetic , Transcription Factors/metabolism , Tumor Suppressor Proteins , 3T3 Cells , Animals , Binding Sites , Biomarkers, Tumor , Mice , Upstream Stimulatory Factors
5.
Mol Cell Biol ; 10(2): 653-61, 1990 Feb.
Article in English | MEDLINE | ID: mdl-2300058

ABSTRACT

We have identified a sequence element that specifies the position of transcription initiation for the dihydrofolate reductase gene. Unlike the functionally analogous TATA box that directs RNA polymerase II to initiate transcription 30 nucleotides downstream, the positioning element of the dihydrofolate reductase promoter is located directly at the site of transcription initiation. By using DNase I footprint analysis, we have shown that a protein binds to this initiator element. Transcription initiated at the dihydrofolate reductase initiator element when 28 nucleotides were inserted between it and all other upstream sequences, or when it was placed on either side of the DNA helix, suggesting that there is no strict spatial requirement between the initiator and an upstream element. Although neither a single Sp1-binding site nor a single initiator element was sufficient for transcriptional activity, the combination of one Sp1-binding site and the dihydrofolate reductase initiator element cloned into a plasmid vector resulted in transcription starting at the initiator element. We have also shown that the simian virus 40 late major initiation site has striking sequence homology to the dihydrofolate reductase initiation site and that the same, or a similar, protein binds to both sites. Examination of the sequences at other RNA polymerase II initiation sites suggests that we have identified an element that is important in the transcription of other housekeeping genes. We have thus named the protein that binds to the initiator element HIP1 (Housekeeping Initiator Protein 1).


Subject(s)
DNA-Binding Proteins/metabolism , Genes , Promoter Regions, Genetic , RNA Polymerase II/metabolism , Tetrahydrofolate Dehydrogenase/genetics , Transcription Factors/metabolism , Transcription, Genetic , Base Sequence , Binding Sites , Deoxyribonuclease I , HeLa Cells/enzymology , Humans , Molecular Sequence Data , Oligonucleotide Probes , Plasmids , Protein Binding , Sequence Homology, Nucleic Acid
6.
Mol Cell Biol ; 10(7): 3788-91, 1990 Jul.
Article in English | MEDLINE | ID: mdl-2113176

ABSTRACT

The Krox-20 gene is rapidly and transiently induced when quiescent 3T3 cells are stimulated to reenter the proliferative cycle. We identified the major serum-responsive transcription initiation site and found that it differs from the initiation sites previously identified for the Krox-20 gene. Transcripts from the major serum-responsive initiation site increased at least 40-fold in serum-stimulated cells compared with logarithmically growing cells.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation , Metalloproteins/genetics , Transcription Factors/genetics , Transcription, Genetic , Animals , Base Sequence , Blood , Cells, Cultured , Culture Media , Cycloheximide/pharmacology , Early Growth Response Protein 2 , Genes , Kinetics , Mice , Molecular Sequence Data , Nucleotide Mapping , RNA, Messenger/genetics , Sequence Homology, Nucleic Acid , Transcription, Genetic/drug effects
7.
Mol Cell Biol ; 10(4): 1390-8, 1990 Apr.
Article in English | MEDLINE | ID: mdl-2320003

ABSTRACT

The murine dihydrofolate reductase gene is regulated by a bidirectional promoter that lacks a TATA box. To identify the DNA sequences required for dihydrofolate reductase transcription, the activities of various templates were determined by in vitro transcription analysis. Our data indicate that sequences both upstream and downstream of the transcription initiation site modulate the activity of the dihydrofolate reductase promoter. We have focused on two regions downstream of the transcription initiation site that are important in determining the overall efficiency of the promoter. Region 1, which included exon 1 and part of intron 1, could stimulate transcription when placed in either orientation in the normal downstream position and when inserted upstream of the transcription start site. This region could also stimulate transcription in trans when the enhancer was physically separate from the promoter. Deletion of region 2, spanning 46 nucleotides of the 5' untranslated region, reduced transcriptional activity by fivefold. DNase I footprinting reactions identified protein-binding sites in both downstream stimulatory regions. Protein bound to two sites in region 1, both of which contain an inverted CCAAT box. The protein-binding site in the 5' untranslated region has extensive homology to binding sites in promoters that both lack (simian virus 40 late) and contain (adenovirus type 2 major late promoter and c-myc) TATA boxes.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation, Enzymologic , Promoter Regions, Genetic , Tetrahydrofolate Dehydrogenase/genetics , Transcription, Genetic , Animals , Base Sequence , Cell Line , Cell Nucleus/metabolism , Deoxyribonuclease I , Exons , HeLa Cells/metabolism , Humans , Introns , Mice , Molecular Sequence Data , Plasmids , Restriction Mapping , Sequence Homology, Nucleic Acid , Teratoma
8.
Mol Cell Biol ; 9(10): 4568-70, 1989 Oct.
Article in English | MEDLINE | ID: mdl-2479829

ABSTRACT

In vitro reactions identified a transcription initiation site located 740 nucleotides upstream of the dihydrofolate reductase translational start. Transcription from this site proceeded in the direction opposite to that of dihydrofolate reductase mRNA. Deletion mapping indicated that this new promoter can be separated from the dihydrofolate reductase promoter and that separation increased transcription at -740. Transcripts that initiate at -740 were also detected in cellular RNA, indicating that this is a bona fide transcription initiation site in vivo.


Subject(s)
Promoter Regions, Genetic/genetics , Tetrahydrofolate Dehydrogenase/genetics , Animals , Cell Nucleus/analysis , Cytoplasm/analysis , DNA Mutational Analysis , Mice , RNA/analysis , Transcription, Genetic
9.
Mol Cell Biol ; 15(5): 2527-35, 1995 May.
Article in English | MEDLINE | ID: mdl-7739536

ABSTRACT

To better understand the signaling pathways which lead to DNA synthesis in mammalian cells, we have studied the transcriptional activation of genes needed during the S phase of the cell cycle. Transcription of the gene encoding a pyrimidine biosynthetic enzyme, carbamoyl-phosphate synthase (glutamine-hydrolyzing)/aspartate carbamoyltransferase/dihydroorotase (cad), increases at the G1/S-phase boundary. We have mapped the growth-dependent response element in the hamster cad gene to the extended palindromic E-box sequence, CCACGTGG, which is centered at +65 in the 5' untranslated sequence. Mutation of the E box abolished growth-dependent transcription, and an oligonucleotide corresponding to the cad sequence at +55 to +75 (+55/+75) restored growth-dependent regulation to nonresponsive cad promoter mutants when placed down-stream of the transcription start site. The same oligonucleotide conferred less G1/S-phase induction when placed upstream of basal promoter elements. An analogous oligonucleotide containing the mutant E box had no effect in either location. Nuclear proteins bound the cad +55/+75 element in a cell cycle-dependent manner in electromobility shift assays; antibodies specific to USF and Max blocked the DNA-binding activity of different growth-regulated protein-DNA complexes. Expression of c-Myc mutants which have been shown to dominantly interfere with the function of c-Myc and Max significantly inhibited cad transcription during S phase but had no effect on transcription from another G1/S-phase-activated promoter, dhfr. These data support a model whereby E-box-binding proteins activate serum-induced transcription from the cad promoter at the G1/S-phase boundary and suggest that a Max-associated protein complex contributes to the serum response.


Subject(s)
Aspartate Carbamoyltransferase/genetics , Carbamoyl-Phosphate Synthase (Glutamine-Hydrolyzing)/genetics , Dihydroorotase/genetics , Genes, myc , Multienzyme Complexes/genetics , 3T3 Cells , Animals , Base Sequence , Binding Sites/genetics , Cricetinae , DNA/genetics , DNA/metabolism , G1 Phase/genetics , Mesocricetus , Mice , Molecular Sequence Data , Mutation , Promoter Regions, Genetic , S Phase/genetics , Suppression, Genetic , Transcription, Genetic
10.
Mol Cell Biol ; 17(4): 1966-76, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9121444

ABSTRACT

Activity of the dihydrofolate reductase (dhfr) promoter increases at the G1-S-phase boundary of the cell cycle. Mutations that abolish protein binding to an E2F element in the dhfr promoter also abolish the G1-S-phase increase in dhfr transcription, indicating that transcriptional regulation is mediated by the E2F family of proteins. To investigate the mechanism by which E2F regulates dhfr transcription, we moved the E2F element upstream and downstream of its natural position in the promoter. We found that the E2F element confers growth regulation to the dhfr promoter only when it is proximal to the transcription start site. Using a heterologous E2F element, we showed that position-dependent regulation is a property that is promoter specific, not E2F element specific. We demonstrated that E2F-mediated growth regulation of dhfr transcription requires activation of the dhfr promoter in S phase and that the C-terminal activation domains of E2F1, E2F4, and E2F5, when fused to the Gal4 DNA binding domain, are sufficient to specify position-dependent activation. To further investigate the role of activation in dhfr regulation, we tested other transactivation domains for their ability to activate the dhfr promoter. We found that the N-terminal transactivation domain of VP16 cannot activate the dhfr promoter. We propose that, unlike other E2F-regulated promoters, robust transcription from the dhfr promoter requires an E2F transactivation domain close to the transcription start site.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , DNA-Binding Proteins , Promoter Regions, Genetic , Tetrahydrofolate Dehydrogenase/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , 3T3 Cells , Animals , Base Sequence , Binding Sites/genetics , Cloning, Molecular , DNA/genetics , DNA/metabolism , E2F Transcription Factors , E2F1 Transcription Factor , E2F4 Transcription Factor , E2F5 Transcription Factor , HeLa Cells , Humans , Mice , Models, Genetic , Mutation , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1 , Transcription, Genetic , Transcriptional Activation
11.
Mol Cell Biol ; 13(3): 1610-8, 1993 Mar.
Article in English | MEDLINE | ID: mdl-8441401

ABSTRACT

Enhanced expression of genes involved in nucleotide biosynthesis, such as dihydrofolate reductase (DHFR), is a hallmark of entrance into the DNA synthesis (S) phase of the mammalian cell cycle. To investigate the regulated expression of the DHFR gene, we stimulated serum-starved NIH 3T3 cells to synchronously reenter the cell cycle. Our previous results show that a cis-acting element at the site of DHFR transcription initiation is necessary for serum regulation. Recently, this element has been demonstrated to bind the cloned transcription factor E2F. In this study, we focused on the role of E2F in the growth regulation of DHFR. We demonstrated that a single E2F site, in the absence or presence of other promoter elements, was sufficient for growth-regulated promoter activity. Next, we showed that the increase in DHFR mRNA at the G1/S-phase boundary required protein synthesis, raising the possibility that a protein(s) lacking in serum-starved cells is required for DHFR transcription. We found that, similar to DHFR mRNA expression, levels of murine E2F1 mRNA were low in serum-starved cells and increased at the G1/S-phase boundary in a protein synthesis-dependent manner. Furthermore, in a cotransfection experiment, expression of human E2F1 stimulated the DHFR promoter 22-fold in serum-starved cells. We suggest that E2F1 may be the key protein required for DHFR transcription that is absent in serum-starved cells. Expression of E2F also abolished the serum-stimulated regulation of the DHFR promoter and resulted in transcription patterns similar to those seen with expression of the adenoviral oncoprotein E1A. In summary, we provide evidence for the importance of E2F in the growth regulation of DHFR and suggest that alterations in the levels of E2F may have severe consequences in the control of cellular proliferation.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , Gene Expression Regulation, Enzymologic , Promoter Regions, Genetic/genetics , RNA, Messenger/biosynthesis , Tetrahydrofolate Dehydrogenase/genetics , Transcription Factors/genetics , 3T3 Cells , Animals , Base Sequence , Cell Division/drug effects , Culture Media, Serum-Free , DNA-Binding Proteins/genetics , E2F Transcription Factors , E2F1 Transcription Factor , G1 Phase , Mice , Molecular Sequence Data , Nuclear Proteins/genetics , Protein Biosynthesis , Retinoblastoma-Binding Protein 1 , S Phase , Serum Response Factor , Transcription Factor DP1
12.
Mol Cell Biol ; 14(3): 1861-9, 1994 Mar.
Article in English | MEDLINE | ID: mdl-8114719

ABSTRACT

E2F has been implicated in growth control because of its association with the retinoblastoma protein and the presence of E2F binding sites in the promoters of several growth-regulated genes. Proteins that bind to an E2F site have been cloned from human and mouse cells. However, these two proteins (human E2F1 and mouse DP-1) are quite different in sequence. We have now cloned a mouse cDNA encoding a protein 86% identical to the human E2F1 protein. The mouse E2F1 cDNA encodes a 430-amino-acid protein with a predicted molecular weight of 46,322 and detects mRNAs of 2.7 and 2.2 kb. Using primers complementary to sequences in the mouse E2F1 3' untranslated region, we mapped the mouse E2F1 gene to chromosome 2, near the Agouti and c-src loci. To understand the role of the different E2F family members in the growth of mouse NIH 3T3 cells, we examined the levels of E2F1 and DP-1 mRNAs in different stages of the cell cycle. Since the levels of E2F1 but not DP-1 mRNA correlated with changes in transcription from the dhfr promoter, we examined whether E2F1 could activate various growth-regulated promoters. We found that E2F1 could activate some (dhfr, thymidine kinase, and DNA polymerase alpha) but not all (thymidylate synthase, cad, and c-myc) of these promoters. On the basis of changes in levels of E2F1 and its ability to transactivate growth-regulated promoters, we propose that E2F1 may mediate growth factor-initiated signal transduction.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , Transcription Factors/genetics , Amino Acid Sequence , Animals , Base Sequence , Cell Cycle , Chromosome Mapping , Cloning, Molecular , DNA, Complementary/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , E2F Transcription Factors , E2F1 Transcription Factor , Gene Expression Regulation , Genes , Mice , Molecular Sequence Data , Promoter Regions, Genetic , RNA, Messenger/genetics , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1 , Transcription, Genetic
13.
Mol Cell Biol ; 21(20): 6820-32, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11564866

ABSTRACT

We have taken a new approach to the identification of E2F-regulated promoters. After modification of a chromatin immunoprecipitation assay, we cloned nine chromatin fragments which represent both strong and weak in vivo E2F binding sites. Further characterization of three of the cloned fragments revealed that they are bound in vivo not only by E2Fs but also by members of the retinoblastoma tumor suppressor protein family and by RNA polymerase II, suggesting that these fragments represent promoters regulated by E2F transcription complexes. In fact, database analysis indicates that all three fragments correspond to genomic DNA located just upstream of start sites for previously identified mRNAs. One clone, ChET 4, corresponds to the promoter region for beclin 1, a candidate tumor suppressor protein. We demonstrate that another of the clones, ChET 8, is strongly bound by E2F family members in vivo but does not contain a consensus E2F binding site. However, this fragment functions as a promoter whose activity can be repressed by E2F1. Finally, we demonstrate that the ChET 9 promoter contains a consensus E2F binding site, can be activated by E2F1, and drives expression of an mRNA that is upregulated in colon and liver tumors. Interestingly, the characterized ChET promoters do not display regulation patterns typical of known E2F target genes in a U937 cell differentiation system. In summary, we have provided evidence that chromatin immunoprecipitation can be used to identify E2F-regulated promoters which contain both consensus and nonconsensus binding sites and have shown that not all E2F-regulated promoters show identical expression profiles.


Subject(s)
Cell Cycle Proteins , Chromatin/metabolism , Cloning, Molecular , DNA-Binding Proteins , Neoplasm Proteins , Precipitin Tests/methods , Promoter Regions, Genetic , Transcription Factors/genetics , Apoptosis Regulatory Proteins , Beclin-1 , Binding Sites , Cell Differentiation , E2F Transcription Factors , E2F1 Transcription Factor , Exons , HeLa Cells , Homeodomain Proteins/genetics , Humans , Membrane Proteins , Models, Genetic , Polymerase Chain Reaction , Protein Binding , Proteins/genetics , Proto-Oncogene Proteins c-myc/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/chemistry , Transfection , U937 Cells , Up-Regulation
14.
Mol Cell Biol ; 12(3): 1054-63, 1992 Mar.
Article in English | MEDLINE | ID: mdl-1545788

ABSTRACT

The transcription rate of the dihydrofolate reductase (DHFR) gene increases at the G1/S boundary of the proliferative cell cycle. Through analysis of transiently and stably transfected NIH 3T3 cells, we have now demonstrated that DHFR promoter sequences extending from -270 to +20 are sufficient to confer similar regulation on a reporter gene. Mutation of a protein binding site that spans sequences from -16 to +11 in the DHFR promoter resulted in loss of the transcriptional increase at the G1/S boundary. Purification of an activity from HeLa nuclear extract that binds to this region enriched for a 180-kDa polypeptide (HIP1). Using this HIP1 preparation, we have identified specific positions within the binding site that are critical for efficient protein-DNA interactions. An analysis of association and dissociation rates suggests that bound HIP1 protein can exchange rapidly with free protein. This rapid exchange may facilitate the burst of transcriptional activity from the DHFR promoter at the G1/S boundary.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation, Enzymologic , Promoter Regions, Genetic , Tetrahydrofolate Dehydrogenase/genetics , 3T3 Cells , Animals , Base Sequence , Binding Sites , Cell Division/genetics , DNA/metabolism , DNA-Binding Proteins/isolation & purification , Mice , Molecular Sequence Data , Molecular Weight , Mutagenesis, Site-Directed , Transfection
15.
Mol Cell Biol ; 12(5): 2250-9, 1992 May.
Article in English | MEDLINE | ID: mdl-1569952

ABSTRACT

We examined the ability of purified RNA polymerase (RNAP) II lacking the carboxy-terminal heptapeptide repeat domain (CTD), called RNAP IIB, to transcribe a variety of promoters in HeLa extracts in which endogenous RNAP II activity was inhibited with anti-CTD monoclonal antibodies. Not all promoters were efficiently transcribed by RNAP IIB, and transcription did not correlate with the in vitro strength of the promoter or with the presence of a consensus TATA box. This was best illustrated by the GC-rich, non-TATA box promoters of the bidirectional dihydrofolate reductase (DHFR)-REP-encoding locus. Whereas the REP promoter was transcribed by RNAP IIB, the DHFR promoter remained inactive after addition of RNAP IIB to the antibody-inhibited reactions. However, both promoters were efficiently transcribed when purified RNAP with an intact CTD was added. We analyzed a series of promoter deletions to identify which cis elements determine the requirement for the CTD of RNAP II. All of the promoter deletions of both DHFR and REP retained the characteristics of their respective full-length promoters, suggesting that the information necessary to specify the requirement for the CTD is contained within approximately 65 bp near the initiation site. Furthermore, a synthetic minimal promoter of DHFR, consisting of a single binding site for Sp1 and a binding site for the HIP1 initiator cloned into a bacterial vector sequence, required RNAP II with an intact CTD for activity in vitro. Since the synthetic minimal promoter of DHFR and the smallest REP promoter deletion are both activated by Sp1, the differential response in this assay does not result from upstream activators. However, the sequences around the start sites of DHFR and REP are not similar and our data suggest that they bind different proteins. Therefore, we propose that specific initiator elements are important for determination of the requirement of some promoters for the CTD.


Subject(s)
Cell Nucleus/enzymology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Promoter Regions, Genetic , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Tetrahydrofolate Dehydrogenase/genetics , Thymus Gland/enzymology , Transcription, Genetic , Animals , Base Sequence , Binding Sites , Cattle , Chromosome Deletion , HeLa Cells , Humans , Kinetics , Molecular Sequence Data , Plasmids , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-raf , RNA Polymerase II/isolation & purification , Restriction Mapping , Templates, Genetic
16.
Mol Cell Biol ; 20(16): 5797-807, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10913163

ABSTRACT

E2F-mediated transcription is thought to involve binding of an E2F-pocket protein complex to promoters in the G(0) phase of the cell cycle and release of the pocket protein in late G(1), followed by release of E2F in S phase. We have tested this model by monitoring protein-DNA interactions in living cells using a formaldehyde cross-linking and immunoprecipitation assay. We find that E2F target genes are bound by distinct E2F-pocket protein complexes which change as cells progress through the cell cycle. We also find that certain E2F target gene promoters are bound by pocket proteins when such promoters are transcriptionally active. Our data indicate that the current model applies only to certain E2F target genes and suggest that Rb family members may regulate transcription in both G(0) and S phases. Finally, we find that a given promoter can be bound by one of several different E2F-pocket protein complexes at a given time in the cell cycle, suggesting that cell cycle-regulated transcription is a stochastic, not a predetermined, process.


Subject(s)
Carrier Proteins , Cell Cycle Proteins/genetics , Gene Expression Regulation , Nuclear Proteins/genetics , Transcription Factors/genetics , Transcription, Genetic , 3T3 Cells , Animals , Cell Cycle/genetics , DNA-Binding Proteins/genetics , E2F Transcription Factors , Mice , Promoter Regions, Genetic , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1
17.
Mol Cell Biol ; 21(17): 6006-16, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11486038

ABSTRACT

We show that the Mre11 complex associates with E2F family members via the Nbs1 N terminus. This association and Nbs1 phosphorylation are correlated with S-phase checkpoint proficiency, whereas neither is sufficient individually for checkpoint activation. The Nbs1 E2F interaction occurred near the Epstein-Barr virus origin of replication as well as near a chromosomal replication origin in the c-myc promoter region and was restricted to S-phase cells. The Mre11 complex colocalized with PCNA at replication forks throughout S phase, both prior to and coincident with the appearance of nascent DNA. These data suggest that the Mre11 complex suppresses genomic instability through its influence on both the regulation and progression of DNA replication.


Subject(s)
Cell Cycle Proteins , DNA Replication , DNA-Binding Proteins/metabolism , Transcription Factors/metabolism , Animals , Binding Sites , Cell Line , DNA Repair Enzymes , E2F Transcription Factors , HeLa Cells , Humans , MRE11 Homologue Protein , Mice , Nuclear Proteins/metabolism , Phosphorylation , S Phase , Signal Transduction , Tumor Cells, Cultured
18.
Oncogene ; 19(18): 2257-68, 2000 Apr 27.
Article in English | MEDLINE | ID: mdl-10822376

ABSTRACT

To gain insight into the tumor suppressor properties of E2F1, we investigated growth inhibition by the E2F family of transcription factors using a tissue culture model system. We first show that exogenous E2F expression causes an 80% decrease in NIH3T3 colony formation and activated c-Ha-Ras-mediated focus formation. Inhibition of Ras-mediated transformation was dependent upon E2F DNA binding activity but did not require amino- or carboxy-terminal E2F1 protein interaction domains. Because E2F upregulation has been suggested to be associated with a neoplastic phenotype, it was possible that increased E2F activity would not be inhibitory to previously transformed cells. However, we found that exogenous E2F was also inhibitory to growth of NIH3T3 cells previously transformed by Ras or Neu. Further characterization revealed that exogenous E2F expression is inhibitory at very early times after transfection, causing dramatic losses in transfected cell populations. Interestingly, those few cells which do establish appear to be unaffected by the overexpressed E2F. Therefore, we propose that increased E2F activity may only be tolerated in a subset of cells which have acquired specific alterations that are dominant over E2F-mediated growth inhibition.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , Cell Transformation, Neoplastic/genetics , DNA-Binding Proteins , Genes, Tumor Suppressor , Genes, erbB-2 , Genes, ras , Transcription Factors/genetics , 3T3 Cells , Animals , Colony-Forming Units Assay , E2F Transcription Factors , E2F1 Transcription Factor , Mice , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1
19.
Oncogene ; 20(21): 2704-12, 2001 May 10.
Article in English | MEDLINE | ID: mdl-11420682

ABSTRACT

Liver cancer is the fifth most common cancer worldwide and unlike certain other cancers, such as colon cancer, a mutational model has not yet been developed. We have performed gene expression profiling of normal and neoplastic livers in C3H/HeJ mice treated with diethylnitrosamine. Using oligonucleotide microarrays, we compared gene expression in liver tumors to three different states of the normal liver: quiescent adult, regenerating adult, and newborn. Although each comparison revealed hundreds of differentially expressed genes, only 22 genes were found to be deregulated in the tumors in all three comparisons. Three of these genes were examined in human hepatocellular carcinomas and were found to be upregulated. As a second method of analysis, we used Representational Difference Analysis (RDA) to clone mRNA fragments differentially expressed in liver tumors versus regenerating livers. We cloned several novel mRNAs that are differentially regulated in murine liver tumors. Here we report the sequence of a novel cDNA whose expression is upregulated in both murine and human hepatocellular carcinomas. Our results suggest that DEN-treated mice provide an excellent model for human hepatocellular carcinomas.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms, Experimental/genetics , Liver Neoplasms/genetics , Amino Acid Sequence , Animals , Carcinoma, Hepatocellular/metabolism , Cloning, Molecular/methods , DNA, Complementary/genetics , DNA, Complementary/isolation & purification , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/metabolism , Liver Neoplasms, Experimental/metabolism , Liver Regeneration/genetics , Male , Mice , Mice, Inbred C3H , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
20.
J Mol Biol ; 309(1): 99-120, 2001 May 25.
Article in English | MEDLINE | ID: mdl-11491305

ABSTRACT

The processes that take place during development and differentiation are directed through coordinated regulation of expression of a large number of genes. One such gene regulatory network provides cell cycle control in eukaryotic organisms. In this work, we have studied the structural features of the 5' regulatory regions of cell cycle-related genes. We developed a new method for identifying composite substructures (modules) in regulatory regions of genes consisting of a binding site for a key transcription factor and additional contextual motifs: potential targets for other transcription factors that may synergistically regulate gene transcription. Applying this method to cell cycle-related promoters, we created a program for context-specific identification of binding sites for transcription factors of the E2F family which are key regulators of the cell cycle. We found that E2F composite modules are found at a high frequency and in close proximity to the start of transcription in cell cycle-related promoters in comparison with other promoters. Using this information, we then searched for E2F sites in genomic sequences with the goal of identifying new genes which play important roles in controlling cell proliferation, differentiation and apoptosis. Using a chromatin immunoprecipitation assay, we then experimentally verified the binding of E2F in vivo to the promoters predicted by the computer-assisted methods. Our identification of new E2F target genes provides new insight into gene regulatory networks and provides a framework for continued analysis of the role of contextual promoter features in transcriptional regulation. The tools described are available at http://compel.bionet.nsc.ru/FunSite/SiteScan.html.


Subject(s)
Cell Cycle Proteins , Cell Cycle/genetics , Computational Biology/methods , DNA-Binding Proteins , Gene Expression Regulation , Genes, cdc , Response Elements/genetics , Transcription Factors/metabolism , Animals , Base Sequence , Binding Sites , Chromatin/genetics , Chromatin/metabolism , Cross-Linking Reagents , Databases as Topic , E2F Transcription Factors , Formaldehyde , Gene Frequency , Humans , Internet , Phosphoproteins/genetics , Phylogeny , Precipitin Tests , Promoter Regions, Genetic/genetics , RNA-Binding Proteins/genetics , Reproducibility of Results , Sensitivity and Specificity , Software , Transcription, Genetic/genetics , Nucleolin
SELECTION OF CITATIONS
SEARCH DETAIL