Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Methods ; 22(2): 116-9, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11020325

ABSTRACT

Analyzing mutation spectra is a very powerful method to determine the effects of various types of DNA damage and to understand the workings of various DNA repair pathways. However, compiling sequence-specific mutation spectra is laborious; even with modern sequencing technology, it is rare to obtain spectra with more than several hundred data points. Two assay systems are described for yeast, one for insertion/deletion mutations and one for base substitution mutations, that allow determination of specific mutations without the necessity of DNA sequencing. The assay for insertion/deletion mutations uses a variety of different simple repeats placed in frame with URA3 such that insertions or deletions lead to a selectable Ura(-) phenotype; essentially all such mutations are in the simple repeat sequence. The assay for base substitution mutations uses a series of six strains with different mutations in one essential codon of the CYC1 gene. Because only true reversions lead to a selectable phenotype, the bases mutated in any reversion event are known. The advantage of these assays is that they can quantitatively determine over several orders of magnitude the types of mutations that occur under a given set of conditions, without DNA sequencing.


Subject(s)
DNA Mutational Analysis/methods , Mutagenicity Tests/methods , Plasmids/genetics , Yeasts/genetics , DNA Damage , DNA Repair , Gene Deletion , Point Mutation
2.
Genetics ; 154(1): 133-46, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10628975

ABSTRACT

The Saccharomyces cerevisiae homologs of the bacterial mismatch repair proteins MutS and MutL correct replication errors and prevent recombination between homeologous (nonidentical) sequences. Previously, we demonstrated that Msh2p, Msh3p, and Pms1p regulate recombination between 91% identical inverted repeats, and here use the same substrates to show that Mlh1p and Msh6p have important antirecombination roles. In addition, substrates containing defined types of mismatches (base-base mismatches; 1-, 4-, or 12-nt insertion/deletion loops; or 18-nt palindromes) were used to examine recognition of these mismatches in mitotic recombination intermediates. Msh2p was required for recognition of all types of mismatches, whereas Msh6p recognized only base-base mismatches and 1-nt insertion/deletion loops. Msh3p was involved in recognition of the palindrome and all loops, but also had an unexpected antirecombination role when the potential heteroduplex contained only base-base mismatches. In contrast to their similar antimutator roles, Pms1p consistently inhibited recombination to a lesser degree than did Msh2p. In addition to the yeast MutS and MutL homologs, the exonuclease Exo1p and the nucleotide excision repair proteins Rad1p and Rad10p were found to have roles in inhibiting recombination between mismatched substrates.


Subject(s)
Base Pair Mismatch/genetics , DNA Repair/genetics , Mitosis/genetics , Recombination, Genetic , Saccharomyces cerevisiae/genetics , Base Sequence , DNA Primers , Epistasis, Genetic , Repetitive Sequences, Nucleic Acid , Saccharomyces cerevisiae/cytology
3.
Proc Natl Acad Sci U S A ; 95(26): 15487-91, 1998 Dec 22.
Article in English | MEDLINE | ID: mdl-9860995

ABSTRACT

In most organisms, the mismatch repair (MMR) system plays an important role in substantially lowering mutation rates and blocking recombination between nonidentical sequences. In Saccharomyces cerevisiae, the products of three genes homologous to Escherichia coli mutS-MSH2, MSH3, and MSH6-function in MMR by recognizing mispaired bases. To determine the effect of MMR on single-base pair mismatches, we have measured reversion rates of specific point mutations in the CYC1 gene in both wild-type and MMR-deficient strains. The reversion rates of all of the point mutations are similar in wild-type cells. However, we find that in the absence of MSH2 or MSH6, but not MSH3, reversion rates of some mutations are increased by up to 60,000-fold, whereas reversion rates of other mutations are essentially unchanged. When cells are grown anaerobically, the reversion rates in MMR-deficient strains are decreased by as much as a factor of 60. We suggest that the high reversion rates observed in these MMR-deficient strains are caused by misincorporations opposite oxidatively damaged bases and that MMR normally prevents these mutations. We further suggest that recognition of mispairs opposite damaged bases may be a more important role for MMR in yeast than correction of errors opposite normal bases.


Subject(s)
Base Pair Mismatch/genetics , DNA Repair , Point Mutation , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/genetics , Aerobiosis , Anaerobiosis , DNA Damage , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Diploidy , Fungal Proteins/genetics , Fungal Proteins/metabolism , Gene Deletion , MutS Homolog 2 Protein , MutS Homolog 3 Protein , Saccharomyces cerevisiae/growth & development
4.
Cell ; 91(4): 467-77, 1997 Nov 14.
Article in English | MEDLINE | ID: mdl-9390556

ABSTRACT

Mice carrying a null mutation in the mismatch repair gene Msh6 were generated by gene targeting. Cells that were homozygous for the mutation did not produce any detectable MSH6 protein, and extracts prepared from these cells were defective for repair of single nucleotide mismatches. Repair of 1, 2, and 4 nucleotide insertion/deletion mismatches was unaffected. Mice that were homozygous for the mutation had a reduced life span. The mice developed a spectrum of tumors, the most predominant of which were gastrointestinal tumors and B- as well as T-cell lymphomas. The tumors did not show any microsatellite instability. We conclude that MSH6 mutations, like those in some other members of the family of mismatch repair genes, lead to cancer susceptibility, and germline mutations in this gene may be associated with a cancer predisposition syndrome that does not show microsatellite instability.


Subject(s)
DNA Repair/genetics , DNA-Binding Proteins/genetics , Mutation/physiology , Neoplasms/genetics , Adenomatous Polyposis Coli Protein , Animals , Base Sequence , Cell Line , Cloning, Molecular , Cytoskeletal Proteins/analysis , Female , Gastrointestinal Neoplasms/chemistry , Gastrointestinal Neoplasms/genetics , Genes, APC , Genetic Predisposition to Disease , Germ-Line Mutation/physiology , Heterozygote , Homozygote , Lymphoma/genetics , Lymphoma/pathology , Male , Mice , Mice, Mutant Strains , Microsatellite Repeats/genetics , Molecular Sequence Data , Neoplasms/chemistry , Neoplasms/pathology , RNA, Messenger/analysis
5.
Genetics ; 143(3): 1127-35, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8807287

ABSTRACT

Transcription-coupled repair, the targeted repair of the transcribed strands of active genes, is defective in bacteria, yeast, and human cells carrying mutations in mfd, RAD26 and ERCC6, respectively. Other factors probably are also uniquely involved in transcription-repair coupling. Recently, a defect was described in transcription-coupled repair for Escherichia coli mismatch repair mutants and human tumor cell lines with mutations in mismatch repair genes. We examined removal of UV-induced DNA damage in yeast strains mutated in mismatch repair genes in an effort to confirm a defect in transcription-coupled repair in this system. In addition, we determined the contribution of the mismatch repair gene MSH2 to transcription-coupled repair in the absence of global genomic repair using rad7 delta mutants. We also determined whether the Rad26-independent transcription-coupled repair observed in rad26 delta and rad7 delta rad26 delta mutants depends on MSH2 by examining repair deficiencies of rad26 delta msh2 delta and rad7 delta rad26 delta msh2 delta mutants. We found no defects in transcription-coupled repair caused by mutations in the mismatch repair genes MSH2, MLH1, PMS1, and MSH3. Yeast appears to differ from bacteria and human cells in the capacity for transcription-coupled repair in a mismatch repair mutant background.


Subject(s)
Cell Cycle Proteins , DNA Damage , DNA Repair , DNA-Binding Proteins/genetics , Fungal Proteins/genetics , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/genetics , Schizosaccharomyces pombe Proteins , Transcription, Genetic , Gene Deletion , MutS Homolog 2 Protein , Mutation , Saccharomyces cerevisiae/radiation effects , Ultraviolet Rays
6.
Mol Cell Biol ; 16(3): 1085-93, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8622653

ABSTRACT

Mismatch repair systems correct replication- and recombination-associated mispaired bases and influence the stability of simple repeats. These systems thus serve multiple roles in maintaining genetic stability in eukaryotes, and human mismatch repair defects have been associated with hereditary predisposition to cancer. In prokaryotes, mismatch repair systems also have been shown to limit recombination between diverged (homologous) sequences. We have developed a unique intron-based assay system to examine the effects of yeast mismatch repair genes (PMS1, MSH2, and MSH3) on crossovers between homologous sequences. We find that the apparent antirecombination effects of mismatch repair proteins in mitosis are related to the degree of substrate divergence. Defects in mismatch repair can elevate homologous recombination between 91% homologous substrates as much as 100-fold while having only modest effects on recombination between 77% homologous substrates. These observations have implications for genome stability and general mechanisms of recombination in eukaryotes.


Subject(s)
Fungal Proteins/genetics , Genes, Fungal , Saccharomyces cerevisiae/metabolism , Base Sequence , DNA Repair , Mitosis , Molecular Sequence Data , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Sequence Alignment , Sequence Analysis
7.
Gene ; 169(1): 111-3, 1996 Feb 22.
Article in English | MEDLINE | ID: mdl-8635733

ABSTRACT

Cassettes based on a hisG-URA3-hisG insert have been modified by the addition of an KmR-encoding gene and flanking polylinker sites, greatly simplifying construction of gene disruption vectors in Escherichia coli. After gene disruption in yeast, URA3 can then be excised by recombination between the hisG repeats flanking the gene, permitting reuse of the URA3 marker.


Subject(s)
Genetic Vectors , Mutagenesis, Insertional/methods , Saccharomyces cerevisiae/genetics , Cloning, Molecular/methods , Kanamycin Resistance , Molecular Sequence Data , Recombination, Genetic
8.
Proc Natl Acad Sci U S A ; 92(22): 10418-21, 1995 Oct 24.
Article in English | MEDLINE | ID: mdl-7479796

ABSTRACT

Eukaryotic genomes contain tracts of DNA in which a single base or a small number of bases are repeated (microsatellites). Mutations in the yeast DNA mismatch repair genes MSH2, PMS1, and MLH1 increase the frequency of mutations for normal DNA sequences and destabilize microsatellites. Mutations of human homologs of MSH2, PMS1, and MLH1 also cause microsatellite instability and result in certain types of cancer. We find that a mutation in the yeast gene MSH3 that does not substantially affect the rate of spontaneous mutations at several loci increases microsatellite instability about 40-fold, preferentially causing deletions. We suggest that MSH3 has different substrate specificities than the other mismatch repair proteins and that the human MSH3 homolog (MRP1) may be mutated in some tumors with microsatellite instability.


Subject(s)
DNA-Binding Proteins , Fungal Proteins/genetics , Genes, Fungal , Multidrug Resistance-Associated Proteins , Repetitive Sequences, Nucleic Acid , Sequence Deletion , Base Sequence , DNA Primers , DNA, Satellite/genetics , Fungal Proteins/biosynthesis , Humans , Molecular Sequence Data , MutS Homolog 3 Protein , Mutagenesis , Mutation , Neoplasms/genetics , Polymerase Chain Reaction , Proteins/genetics , Sequence Homology, Nucleic Acid
9.
Genetics ; 139(3): 1175-88, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7768431

ABSTRACT

A homeologous mitotic recombination assay was used to test the role of Saccharomyces cerevisiae mismatch repair genes PMS1, MSH2 and MSH3 on recombination fidelity. A homeologous gene pair consisting of S. cerevisiae SPT15 and its S. pombe homolog were present as a direct repeat on chromosome V, with the exogenous S. pombe sequences inserted either upstream or downstream of the endogenous S. cerevisiae gene. Each gene carried a different inactivating mutation, rendering the starting strain Spt15-. Recombinants that regenerated SPT15 function were scored after nonselective growth of the cells. In strains wild type for mismatch repair, homeologous recombination was depressed 150- to 180-fold relative to homologous controls, indicating that recombination between diverged sequences is inhibited. In one orientation of the homeologous gene pair, msh2 or msh3 mutations resulted in 17- and 9.6-fold elevations in recombination and the msh2 msh3 double mutant exhibited an 43-fold increase, implying that each MSH gene can function independently in trans to prevent homeologous recombination. Homologous recombination was not significantly affected by the msh mutations. In the other orientation, only msh2 strains were elevated (12-fold) for homeologous recombination. A mutation in MSH3 did not affect the rate of recombination in this orientation. Surprisingly, a pms1 deletion mutant did not exhibit elevated homeologous recombination.


Subject(s)
DNA Repair/genetics , Recombination, Genetic/genetics , Saccharomyces cerevisiae/genetics , Base Sequence , Chromosomes/genetics , Chromosomes/metabolism , DNA-Binding Proteins/genetics , Gene Deletion , Mitosis/genetics , Molecular Sequence Data , Mutation , Sequence Homology, Nucleic Acid , TATA-Box Binding Protein , Transcription Factors/genetics
10.
Gene ; 147(2): 169-77, 1994 Sep 30.
Article in English | MEDLINE | ID: mdl-7926796

ABSTRACT

The mouse Rep-3 gene is transcribed divergently from the same promoter region as the dihydrofolate reductase-encoding gene and has a deduced amino-acid sequence that shares identity with the bacterial protein, MutS, which is involved in DNA mismatch repair. We have cloned Rep-3, mapped it and sequenced all of the known exons and their intron junction sequences. We find that the open reading frame is considerably larger than initially reported and that the most abundant form of Rep-3 mRNA encodes a protein of 123 kDa. The gene spans at least 134 kb and consists of 26 exons, including several alternatively spliced exons. All of the exon/intron junctions match the expected consensus sequences with the exception of the splice junctions for intron 6, which has AT and AC dinucleotides instead of the usual GT and AG bordering the exon sequences. The junction sequences for this intron share consensus sequences with three intron sequences from other genes, thereby helping to establish an alternative consensus sequence.


Subject(s)
DNA Repair , Proteins/genetics , Amino Acid Sequence , Animals , Bacterial Proteins/genetics , Base Sequence , DNA , Exons , Fungal Proteins/genetics , Mice , Mice, Inbred C57BL , Molecular Sequence Data , MutS Homolog 3 Protein , Nucleic Acid Heteroduplexes , RNA Splicing
11.
Mol Gen Genet ; 239(1-2): 97-108, 1993 May.
Article in English | MEDLINE | ID: mdl-8510668

ABSTRACT

We have identified a gene in Saccharomyces cerevisiae, MSH3, whose predicted protein product shares extensive sequence similarity with bacterial proteins involved in DNA mismatch repair as well as with the predicted protein product of the Rep-3 gene of mouse. MSH3 was obtained by performing a polymerase chain reaction on yeast genomic DNA using degenerate oligonucleotide primers designed to anneal with the most conserved regions of a gene that would be homologous to Rep-3 and Salmonella typhimurium mutS. MSH3 seems to play some role in DNA mismatch repair, inasmuch as its inactivation results in an increase in reversion rates of two different mutations and also causes an increase in postmeiotic segregation. However, the effect of MSH3 disruption on reversion rates and postmeiotic segregation appears to be much less than that of previously characterized yeast DNA mismatch repair genes. Alignment of the MSH3 sequence with all of the known MutS homologues suggests that its primary function may be different from the role of MutS in repair of replication errors. MSH3 appears to be more closely related to the mouse Rep-3 gene and other similar eukaryotic mutS homologues than to the yeast gene MSH2 and other mutS homologues that are involved in replication repair. We suggest that the primary function of MSH3 may be more closely related to one of the other known functions of mutS, such as its role in preventing recombination between non-identical sequences.


Subject(s)
Adenosine Triphosphatases , Bacterial Proteins/genetics , DNA Repair/genetics , DNA-Binding Proteins , Escherichia coli Proteins , Fungal Proteins/genetics , Genes, Fungal , Saccharomyces cerevisiae/genetics , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , DNA, Fungal , Humans , Meiosis , Methylation , Mitosis/genetics , Molecular Sequence Data , MutS DNA Mismatch-Binding Protein , MutS Homolog 3 Protein , Mutation , Phenotype , Polymerase Chain Reaction , Restriction Mapping , Saccharomyces cerevisiae Proteins , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid
12.
Nucleic Acids Res ; 21(3): 769-70, 1993 Feb 11.
Article in English | MEDLINE | ID: mdl-8441699
13.
Mol Cell Biol ; 10(11): 6003-12, 1990 Nov.
Article in English | MEDLINE | ID: mdl-2233729

ABSTRACT

The mouse dihydrofolate reductase (Dhfr) promoter region is buried within a CpG island (a region rich in unmethylated CpG dinucleotides), has a high G+C content, and lacks CAAT and TATA elements. The region contains four 48-bp repeats, each of which contains an Sp1-binding site. Another gene, Rep-3 (formerly designated Rep-1), shares the same general promoter region with Dhfr, being transcribed in the direction opposite that of Dhfr. Both genes appear to be housekeeping genes and are expressed at relatively low levels in all tissues. The 5' termini of the major Dhfr transcripts are separated from the 5' termini of the Rep-3 transcripts by approximately 140 bp. This curious structural arrangement suggested that the two genes might share common regulatory elements. To investigate the promoter sequences driving bidirectional transcription, a series of promoter mutations was constructed. These mutations were assayed by a replicating minigene system and by promoter fusions to the chloramphenicol acetyltransferase gene. Linker-scanning mutations that spanned the four repeats produced a variety of mRNA transcript phenotypes. The effects were primarily quantitative, generally reducing the abundance of transcripts for one or both genes. Some mutations affected Dhfr in a qualitative manner, such as by changing the startpoint of one of the major Dhfr transcripts or changing the relative abundance of the two major Dhfr transcripts. Additionally, protein transcription factors that bind to sequences in the mouse Dhfr/Rep-3 major promoter region, potentially affecting expression of either or both genes, were investigated by DNase I footprinting. The results indicate that multiple protein-DNA interactions occur in this region, reflecting potentially complex transcriptional control mechanisms that might modulate expression of either or both genes under different physiological conditions.


Subject(s)
Chromosome Deletion , Genes , Promoter Regions, Genetic , Tetrahydrofolate Dehydrogenase/genetics , Animals , Base Sequence , Chloramphenicol O-Acetyltransferase/genetics , Chloramphenicol O-Acetyltransferase/metabolism , Deoxyribonuclease I , Mice , Molecular Sequence Data , Mutation , Plasmids , Transcription, Genetic , Transfection
14.
Gene ; 84(1): 159-64, 1989 Dec 07.
Article in English | MEDLINE | ID: mdl-2691333

ABSTRACT

We demonstrate how a kanamycin-resistance (KmR) cassette flanked by polylinkers with multiple restriction sites can be used to introduce nucleotide (nt) sequence replacements into a region of interest. This method differs in two significant ways from traditional methods of linker mutagenesis. First, the presence of the KmR gene allows for selection of the polylinker, greatly facilitating formation of linker-containing molecules. Second, the polylinker with multiple restriction sites allows a given linker insertion to be combined with a second linker insertion in a variety of different ways and makes possible a range of novel nt to remain in the resulting linker replacement. The result of this flexibility is that fewer different molecules are needed to cover a region, and that relatively large replacements (greater than 40 nt) are possible. We have used this method to introduce a series of sequence replacements that span the mouse dihydrofolate reductase promoter region.


Subject(s)
Escherichia coli/genetics , Kanamycin Resistance/genetics , Mutation , Promoter Regions, Genetic , R Factors , Animals , Base Sequence , Cloning, Molecular/methods , Genes , Mice , Molecular Sequence Data , Restriction Mapping , Tetrahydrofolate Dehydrogenase/genetics , Transcription, Genetic
15.
Gene ; 84(1): 165-72, 1989 Dec 07.
Article in English | MEDLINE | ID: mdl-2691334

ABSTRACT

An intraplasmid recombination system in Escherichia coli has been designed to make possible the engineering of various genes using methods that greatly reduce dependence on appropriately placed restriction enzyme sites. This system has been used to manipulate intervening sequences in dihydrofolate reductase minigenes and to vary the number of 48-bp repeats in the promoter region. In this method, the two fragments to be recombined are cloned into a plasmid separated by a fragment of DNA containing an expressible galactokinase-encoding gene (galK). Selection for loss of the galK gene, but for retention of the plasmid in E. coli, results in a plasmid in which the two fragments have undergone homologous recombination. Several new plasmids are reported here which contain an expressible galK gene flanked by multiple restriction sites. These plasmids should be useful in recombination and as convenient sources of a gene for which both positive and negative selections are available in E. coli.


Subject(s)
Genes , Plasmids , Tetrahydrofolate Dehydrogenase/genetics , Animals , Base Sequence , Cloning, Molecular/methods , DNA, Recombinant/metabolism , Escherichia coli/genetics , Exons , Galactokinase/genetics , Genetic Engineering/methods , Introns , Mice , Molecular Sequence Data , Promoter Regions, Genetic , Restriction Mapping
16.
Mol Cell Biol ; 9(7): 3058-72, 1989 Jul.
Article in English | MEDLINE | ID: mdl-2674679

ABSTRACT

The mouse dihydrofolate reductase gene (dhfr) is a housekeeping gene expressed under the control of a promoter region embedded in a CpG island--a region rich in unmethylated CpG dinucleotides. A divergent transcription unit exists immediately upstream of the dhfr gene which is coamplified with dhfr in some but not all methotrexate-resistant cell lines. We show that the promoter region for this gene pair consists of two bidirectional promoters, a major and minor promoter, which are situated within a 660-base-pair region upstream of the dhfr ATG translation initiation codon. The major promoter controls over 90% of dhfr transcription, while the minor promoter directs the transcription of the remaining dhfr mRNAs. The major promoter functions bidirectionally, transcribing a divergent 4.0-kilobase poly(A) mRNA (class A) in the direction opposite that of dhfr transcription. The predicted protein product of this mRNA is 105 kilodaltons. The minor promoter also functions bidirectionally, directing the transcription of at least two divergent RNAs (class B). These RNAs, present in quantities approximately 1/10 to 1/50 that of the class A mRNAs, are 4.4- and 1.6-kilobase poly(A) mRNAs. cDNAs representing both class A and class B mRNAs have been cloned from a mouse fibroblast cell line which has amplified the dhfr locus (3T3R500). DNA sequence analysis of these cDNAs reveals that the class A and class B mRNAs share, for the most part, the same exons. On the basis of S1 nuclease protection analysis of RNA preparations from several mouse tissues, both dhfr and divergent genes showed similar levels of expression but did show some specificity in start site utilization. Computer homology searches have revealed sequence similarity of the divergent transcripts with bacterial genes involved in DNA mismatch repair, and we therefore have named the divergently transcribed gene Rep-1.


Subject(s)
Cloning, Molecular , Promoter Regions, Genetic , RNA, Messenger/biosynthesis , Tetrahydrofolate Dehydrogenase/genetics , Transcription, Genetic , Amino Acid Sequence , Animals , Bacterial Proteins/genetics , Base Sequence , Cells, Cultured , Chloramphenicol O-Acetyltransferase/genetics , DNA/biosynthesis , DNA/genetics , DNA Repair , Escherichia coli/genetics , Exons , Gene Expression , Immunoblotting , Mice , Molecular Sequence Data , Poly A/genetics , RNA Splicing , RNA, Messenger/classification , RNA, Messenger/genetics , RNA, Messenger/metabolism , Restriction Mapping , Sequence Homology, Nucleic Acid , Tetrahydrofolate Dehydrogenase/biosynthesis
17.
DNA ; 7(10): 729-34, 1988 Dec.
Article in English | MEDLINE | ID: mdl-3069404

ABSTRACT

The use of homologous recombination in Escherichia coli is described as a tool for DNA manipulation. The utility of the method is illustrated by the addition of 3'-flanking sequences to a dhfr minigene by plasmid-phage recombination involving a supF-containing dhfr minigene plasmid and a lambda Charon4A phage containing the 3' end of the dhfr gene. In addition, other uses of both plasmid-phage and phage-phage recombination in gene manipulation are described.


Subject(s)
Escherichia coli/genetics , Bacteriophages/genetics , Chromosome Deletion , Cosmids , Genes, Bacterial , Introns , Plasmids , Recombination, Genetic
18.
Nucleic Acids Res ; 16(14B): 7025-42, 1988 Jul 25.
Article in English | MEDLINE | ID: mdl-3405757

ABSTRACT

We have constructed a plasmid encoding a mouse dihydrofolate reductase (dhfr) minigene which produces dhfr transcripts with all of the 5' and 3' ends observed from the chromosomal mouse dhfr gene. The minigene contains 5' flanking regions, all dhfr coding sequences, one intervening sequence, 11.5 kb of 3' flanking regions beyond the termination codon, an E. coli plasmid origin of replication and antibiotic resistance, and an SV40 minimal origin of replication; the total size is 17.2 kb. When transfected into cells constitutively producing a temperature sensitive SV40 T antigen, the plasmid minigene replicates at the permissive temperature, but fails to replicate at the nonpermissive temperature. Therefore, transcription can be observed in the presence or absence of minigene replication. In addition, a stable divergently transcribed RNA is produced from the dhfr minigene promoter region, with the same 5' ends that are seen in the chromosomal divergently transcribed gene. We show that deletion of the sole remaining intron of the dhfr minigene significantly lowers the amount of dhfr transcript produced but does not affect the amount of divergent transcript. The promoter region for these transcripts contains four 48 bp repeats; reducing the number of these repeats lowers the amount of both dhfr and divergent transcripts produced from the minigene.


Subject(s)
Tetrahydrofolate Dehydrogenase/genetics , Animals , Cell Line , Cloning, Molecular , DNA Replication , Gene Expression Regulation , Genetic Vectors , Introns , Mice , Promoter Regions, Genetic , RNA, Messenger/genetics , Repetitive Sequences, Nucleic Acid , Temperature , Transcription, Genetic
19.
Mol Cell Biol ; 5(8): 1847-58, 1985 Aug.
Article in English | MEDLINE | ID: mdl-3018531

ABSTRACT

The use of murine dihydrofolate reductase (dhfr) gene amplification mutants enabled us to identify important structural and functional features of the dhfr promoter region. We found another transcription unit, at least 14 kilobases in size, which initiates within 130 base pairs of the major dhfr transcript and is transcribed divergently. The 5' ends of both transcripts were analyzed and found to have multiple initiation sites. The major dhfr transcript and the divergent transcript appear to share the same promoter region; the longer transcripts of the dhfr gene overlap with the divergent transcripts and use a different promoter region. The divergent transcript appears to code for a protein; an homologous sequence to its first exon is found in the corresponding location near the human dhfr gene.


Subject(s)
Genes , Promoter Regions, Genetic , Tetrahydrofolate Dehydrogenase/genetics , Transcription, Genetic , Amanitins/pharmacology , Animals , Base Sequence , Cloning, Molecular , Dose-Response Relationship, Radiation , Endonucleases , Gene Amplification , Genes/drug effects , Mice , Mutation , Peptide Chain Initiation, Translational , Single-Strand Specific DNA and RNA Endonucleases , Transcription, Genetic/drug effects , Transcription, Genetic/radiation effects , Ultraviolet Rays
20.
Gene ; 40(1): 151-5, 1985.
Article in English | MEDLINE | ID: mdl-3005123

ABSTRACT

A plasmid, pgam, has been constructed which expresses the phage lambda gene, gam, under the control of the lambda late promoter, p'R, contained in a form of a p'R-qut-t'R1 module. Lambda red- gam-, which normally do not grow on recA- hosts, are able to grow on recA- hosts containing pgam, because their Q function can turn on the gam gene expression. This facilitates cloning with lambda red- gam- vectors in recA- hosts.


Subject(s)
Bacteriophage lambda/genetics , Genes, Viral , Plasmids , DNA Restriction Enzymes , Escherichia coli/genetics , Genes, Bacterial , Genotype , Mutation , Promoter Regions, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...