Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 103
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Genet ; 24(1): 6-8, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10615114

ABSTRACT

The DNA repair picture in humans becomes more complete with the identification of MLH3, a homologue of MutL and a heterodimeric partner of MLH1.


Subject(s)
Base Pair Mismatch , DNA Repair/genetics , Adaptor Proteins, Signal Transducing , Animals , Base Sequence , Carrier Proteins , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , DNA/genetics , Dimerization , Humans , MutL Protein Homolog 1 , Mutation , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nuclear Proteins
2.
Nat Commun ; 13(1): 760, 2022 02 09.
Article in English | MEDLINE | ID: mdl-35140211

ABSTRACT

Prime editing (PE) is a powerful genome engineering approach that enables the introduction of base substitutions, insertions and deletions into any given genomic locus. However, the efficiency of PE varies widely and depends not only on the genomic region targeted, but also on the genetic background of the edited cell. Here, to determine which cellular factors affect PE efficiency, we carry out a focused genetic screen targeting 32 DNA repair factors, spanning all reported repair pathways. We show that, depending on cell line and type of edit, ablation of mismatch repair (MMR) affords a 2-17 fold increase in PE efficiency, across several human cell lines, types of edits and genomic loci. The accumulation of the key MMR factors MLH1 and MSH2 at PE sites argues for direct involvement of MMR in PE control. Our results shed new light on the mechanism of PE and suggest how its efficiency might be optimised.


Subject(s)
DNA Mismatch Repair , Gene Editing , Cell Line , DNA Repair , DNA-Binding Proteins , Genetic Testing , HEK293 Cells , Humans , MutL Protein Homolog 1/genetics , MutS Homolog 2 Protein/metabolism , Mutation, Missense
3.
Science ; 268(5219): 1912-4, 1995 Jun 30.
Article in English | MEDLINE | ID: mdl-7604265

ABSTRACT

DNA mismatch recognition and binding in human cells has been thought to be mediated by the hMSH2 protein. Here it is shown that the mismatch-binding factor consists of two distinct proteins, the 100-kilodalton hMSH2 and a 160-kilodalton polypeptide, GTBP (for G/T binding protein). Sequence analysis identified GTBP as a new member of the MutS homolog family. Both proteins are required for mismatch-specific binding, a result consistent with the finding that tumor-derived cell lines devoid of either protein are also devoid of mismatch-binding activity.


Subject(s)
DNA Repair , DNA, Neoplasm/metabolism , DNA-Binding Proteins/metabolism , Nucleic Acid Heteroduplexes/metabolism , Amino Acid Sequence , Base Composition , Base Sequence , Cloning, Molecular , Colorectal Neoplasms , DNA Repair/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , HeLa Cells , Humans , Molecular Sequence Data , Molecular Weight , Sequence Analysis , Tumor Cells, Cultured
4.
Curr Opin Genet Dev ; 10(2): 157-61, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10753784

ABSTRACT

Post-replicative mismatch repair in humans utilises the hMSH2, hMSH6, hMSH3, hMLH1 and hPMS2 genes and possibly the newly identified hMLH3 gene. Recently, a link has been established between hMSH6 mutations and 'atypical' hereditary non-polyposis colon cancer (HNPCC) with an increased incidence of endometrial cancers. To satisfy the need for a diagnostic test capable of differentiating between pathogenic mutations and polymorphisms, several functional assays that fulfil these criteria have been described. These should allow for better diagnosis of HNPCC.


Subject(s)
Base Pair Mismatch/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , DNA Repair/genetics , Animals , Humans
5.
Curr Biol ; 10(21): R788-90, 2000 Nov 02.
Article in English | MEDLINE | ID: mdl-11084351

ABSTRACT

High-resolution crystal structures have recently been solved for the mismatch binding protein MutS of Escherichia coli and its Thermus aquaticus homologue; they show how these factors recognise such structurally diverse substrates as base-base mismatches and insertion/deletion loops.


Subject(s)
Adenosine Triphosphatases , Bacterial Proteins/chemistry , Base Pair Mismatch , DNA Repair , DNA-Binding Proteins , Escherichia coli Proteins , Escherichia coli/metabolism , Bacterial Proteins/metabolism , Escherichia coli/chemistry , Macromolecular Substances , Models, Molecular , MutS DNA Mismatch-Binding Protein , Protein Structure, Quaternary , Thermus/chemistry , Thermus/metabolism
6.
Curr Biol ; 6(9): 1181-4, 1996 Sep 01.
Article in English | MEDLINE | ID: mdl-8805365

ABSTRACT

In human cells, mismatch recognition is mediated by a heterodimeric complex, hMutSalpha, comprised of two members of the MutS homolog (MSH) family of proteins, hMSH2 and GTBP [1,2]. Correspondingly, tumour-derived cell lines defective in hMSH2 and GTBP have a mutator phenotype [3,4], and extracts prepared from these cells lack mismatch-binding activity [1]. However, although hMSH2 mutant cell lines showed considerable microsatellite instability in tracts of mononucleotide and dinucleotide repeats [4,5], only mononucleotide repeats were somewhat unstable in GTBP mutants [4,6]. These findings, together with data showing that extracts of cells lacking GTBP are partially proficient in the repair of two-nucleotide loops [2], suggested that loop repair can be GTBP-independent. We show here that hMSH2 can also heterodimerize with a third human MSH family member, hMSH3, and that this complex, hMutSbeta, binds loops of one to four extrahelical bases. Our data further suggest that hMSH3 and GTBP are redundant in loop repair, and help explain why only mutations in hMSH2, and not in GTBP or hMSH3, segregate with hereditary non-polyposis colorectal cancer (HNPCC) [7].


Subject(s)
DNA-Binding Proteins/metabolism , DNA/metabolism , Fungal Proteins , Multidrug Resistance-Associated Proteins , Proteins/genetics , Base Sequence , Binding Sites , Biopolymers , DNA-Binding Proteins/genetics , Humans , Molecular Sequence Data , MutS Homolog 2 Protein , MutS Homolog 3 Protein , Mutagenesis, Insertional , Protein Binding , Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Deletion
7.
Curr Biol ; 6(4): 484-6, 1996 Apr 01.
Article in English | MEDLINE | ID: mdl-8723353

ABSTRACT

The process of post-replicative DNA-mismatch repair seems to be highly evolutionarily conserved. In Escherichia coli, DNA mismatches are recognized by the MutS protein. Homologues of the E. coli mutS and mutL mismatch-repair genes have been identified in other prokaryotes, as well as in yeast and mammals. Recombinant Saccharomyces cerevisiae MSH2 (MSH for MutS homologue) and human hMSH2 proteins have been shown to bind to mismatch-containing DNA in vitro. However, the physiological role of hMSH2 is unclear, as shown by the recent finding that the mismatch-binding factor hMutS alpha isolated from extracts of human cells is a heterodimer of hMSH2 and another member of the MSH family, GTBP. It has been reported that S. cerevisiae possesses a mismatch-binding activity, which most probably contains MSH2. We show here that, as in human cells, the S. cerevisiae binding factor is composed of MSH2 and a new functional MutS homologue, MSH6, identified by its homology to GTBP.


Subject(s)
DNA-Binding Proteins/metabolism , Fungal Proteins/metabolism , Saccharomyces cerevisiae Proteins , Amino Acid Sequence , Base Sequence , DNA Repair , DNA, Fungal , HeLa Cells , Humans , Molecular Sequence Data , MutS Homolog 2 Protein , Saccharomyces cerevisiae
8.
Trends Genet ; 10(5): 164-8, 1994 May.
Article in English | MEDLINE | ID: mdl-8036718

ABSTRACT

Recently, both a common inherited human cancer, hereditary non-polyposis colorectal carcinoma (HNPCC), and certain sporadic human cancers have been shown to have frequent alterations in microsatellite sequences. These were ascribed to a defect in the correction of errors of replication; indeed, the cell line H6, which is derived from an HNPCC tumour, has been shown to be deficient in mismatch repair. The HNPCC locus on chromosome 2p has been linked to a gene hMSH2, which encodes a 100 kDa mismatch-binding protein that has an extensive degree of sequence conservation from bacteria through yeast to humans. But could a malfunction of this protein alone cause cancer?


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , DNA Repair , DNA, Neoplasm/genetics , DNA-Binding Proteins/genetics , Proto-Oncogene Proteins/genetics , Amino Acid Sequence , Animals , Humans , Molecular Sequence Data , MutS Homolog 2 Protein
9.
Neoplasma ; 53(4): 269-76, 2006.
Article in English | MEDLINE | ID: mdl-16830052

ABSTRACT

Hereditary non-polyposis colorectal cancer (HNPCC) is associated with germline mutations in DNA mismatch repair genes, predominantly MSH2 and MLH1. Mutation carriers develop cancers in the colorectum, endometrium, ovary, stomach, small intestine and the upper urinary tract. We describe here the results of a mutational analysis of 11 unrelated HNPCC patients by direct genomic sequencing of MLH1 and MSH2. The alterations found include 7 novel changes and 4 different pathogenic mutations described previously in Poland, Moldavia, Finland, Germany, France and USA. Four novel pathogenic mutations in the MLH1 gene include two frameshift mutations (c.1150delG and c.1210_1211delCT), one missense mutation (c.793C>A) and one intron-exon border mutation (c.546- 2A>C). The last change resulted in the skipping of exon 7, as shown by sequencing of RT-PCR products. The only novel MSH2 pathogenic change was a nonsense mutation c.1129C>T. The novel intronic change c.381-41A>G in MLH1 was found in a patient carrying a previously-described mutation in the MSH2 gene. Interestingly, two unrelated patients carried also a novel change in the promoter region of MLH1 in one of the CpG islands (c.-269C>G). However, this alteration does not abrogate transcription, as shown by RT-PCR analysis. In summary, most (approximately 80%) pathogenic germline mutations detected in the studied group of patients by direct genomic sequencing of MLH1 and MSH2 were located in the MLH1 gene. These and previous data indicate that the majority of germline point mutations and small deletions/insertions in HNPCC families in Slovakia affect the MLH1 locus.


Subject(s)
Carrier Proteins/genetics , MutS Homolog 2 Protein/genetics , Nuclear Proteins/genetics , Adaptor Proteins, Signal Transducing , Adult , Base Sequence , Colorectal Neoplasms, Hereditary Nonpolyposis , DNA Repair , Female , Germ-Line Mutation , Humans , Male , Middle Aged , MutL Protein Homolog 1 , Mutation , Slovakia
10.
Nucleic Acids Res ; 29(21): 4378-86, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11691925

ABSTRACT

Bloom's syndrome (BS) is a rare genetic disorder characterised by genomic instability and cancer susceptibility. BLM, the gene mutated in BS, encodes a member of the RecQ family of DNA helicases. Here, we identify hMLH1, which is involved in mismatch repair (MMR) and recombination, as a protein that directly interacts with BLM both in vivo and in vitro, and that the two proteins co-localise to discrete nuclear foci. The interaction between BLM and hMLH1 appears to have been evolutionarily conserved, as Sgs1p, the Saccharomyces cerevisiae homologue of BLM, interacts with yeast Mlh1p. However, cell extracts derived from BS patients show no obvious defects in MMR compared to wild-type- and BLM-complemented BS cell extracts. We conclude that the hMLH1-BLM interaction is not essential for post-replicative MMR, but, more likely, is required for some aspect of genetic recombination.


Subject(s)
Adenosine Triphosphatases/metabolism , Base Pair Mismatch , Bloom Syndrome , DNA Helicases/metabolism , DNA Repair , Neoplasm Proteins/metabolism , Protein Interaction Mapping , Adaptor Proteins, Signal Transducing , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/genetics , Blotting, Far-Western , Carrier Proteins , Cell Line , Cell Nucleus/metabolism , Conserved Sequence , DNA Helicases/chemistry , DNA Helicases/genetics , Fungal Proteins/genetics , Fungal Proteins/metabolism , Humans , MutL Protein Homolog 1 , Mutation/genetics , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Nuclear Proteins/metabolism , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Protein Transport , RecQ Helicases , Recombination, Genetic , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Two-Hybrid System Techniques
11.
Article in English | MEDLINE | ID: mdl-11554300

ABSTRACT

More than 50% of colon cancer-associated mutations in the p53 tumor suppressor gene are C-->T transitions. The majority of them locate in CpG dinucleotides and are thought to have arisen through spontaneous hydrolytic deamination of 5-methylcytosine. This deamination process gives rise to G.T mispairs that need to be repaired to G.C in order to avoid C-->T mutation. Similarly, deamination of cytosine generates G.U mispairs that also produce C-->T transitions if not repaired. Restoration of both G.T and G.U mismatches was shown to be mediated by a short-patch excision repair pathway, and one principal player implicated in this process may be thymine DNA glycosylase (TDG). Human TDG was discovered as an enzyme that has the potential to specifically remove thymine and uracil bases mispaired with guanine through hydrolysis of their N-glycosidic bond, thereby generating abasic sites in DNA and initiating a base excision repair reaction. The same protein was later found to interact physically and functionally with the retinoid receptors RAR and RXR, and this implicated an unexpected function of TDG in nuclear receptor-mediated transcriptional activation of gene expression. The objective of this chapter is to put together the results of different lines of experimentation that have explored the thymine DNA glycosylase since its discovery and to critically evaluate their implications for possible physiological roles of this enzyme.


Subject(s)
Cytosine/analogs & derivatives , DNA Repair , Endodeoxyribonucleases/physiology , Thymine/analogs & derivatives , Thymine/metabolism , Amino Acid Sequence , Animals , Bacterial Proteins/chemistry , Base Pair Mismatch , Base Sequence , Cell Transformation, Neoplastic/genetics , Colonic Neoplasms/genetics , Cytosine/metabolism , DNA Damage , DNA, Neoplasm/genetics , Deamination , Deoxyribonuclease (Pyrimidine Dimer) , Endodeoxyribonucleases/chemistry , Evolution, Molecular , Guanine/chemistry , Humans , Mice , Models, Genetic , Molecular Sequence Data , Receptors, Retinoic Acid/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Structure-Activity Relationship , Substrate Specificity , Thymine/chemistry , Transcription, Genetic , Transfection , Uracil/chemistry
12.
Cancer Res ; 60(12): 3262-70, 2000 Jun 15.
Article in English | MEDLINE | ID: mdl-10866320

ABSTRACT

We investigated the relationship between DNA cytosine methylation and the expression of two genes associated with resistance to DNA methylation damage. Variants of RajiMex- cells acquired resistance to N-methyl-N-nitrosourea by either reactivating a previously silent O6-methylguanine-DNA methyltransferase (MGMT) gene or by repressing the hMSH6 mismatch repair gene. DNA sequencing and measurements of mRNA and enzyme levels revealed that MGMT activity was not correlated with methylation of the core MGMT promoter. Treatment with the demethylating agent 5-azadeoxycytidine reduced MGMT mRNA and enzyme levels, indicating that methylation of some nonpromoter sequences may be required for MGMT gene expression. In contrast, both hMSH6 mRNA and protein levels were increased by 5-azadeoxycytidine treatment of an N-methyl-N-nitrosourea-resistant variant that did not express detectable hMSH6, which implies that this gene was transcriptionally silenced by cytosine methylation. This could be substantiated by in vitro modification of the CpG sites in the hMSH6 promoter with restriction methylase M.SssI, which abolished the transcription of a reporter gene under its control in a transient transfection assay. Taken together, our data show that treatment with chemical methylating agents alters gene expression patterns through increased CpG methylation of genomic DNA, and thereby permits the emergence and selection of clones that are resistant to these agents due to increased repair or tolerance of O6-methylguanine.


Subject(s)
DNA Repair/genetics , DNA-Binding Proteins/genetics , O(6)-Methylguanine-DNA Methyltransferase/genetics , Alkylating Agents/pharmacology , Antimetabolites, Antineoplastic/pharmacology , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Blotting, Western , CpG Islands/genetics , Cytosine/metabolism , DNA Methylation , DNA Repair/drug effects , DNA-Binding Proteins/metabolism , Decitabine , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Gene Silencing , Humans , Luciferases/metabolism , Methylnitrosourea/pharmacology , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Plasmids , Promoter Regions, Genetic , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Transcription, Genetic , Transfection , Tumor Cells, Cultured
13.
Cancer Res ; 61(21): 7719-21, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11691782

ABSTRACT

Conversion of diploidy to haploidy is a method that allows the generation of stable murine/human hybrid cell lines carrying selected human chromosomes in only a single copy. In this setting, it is possible to detect genetic mutations with greater sensitivity and reliability than in diploid cells. Using this method, we were able to identify mutations in the human mismatch repair (MMR) gene hMSH2 in hereditary nonpolyposis colon cancer families, which have escaped detection by the conventional methods. In this report, we show that such hybrid cell lines can also be a valuable tool in the study of the mutated MMR proteins, in particular the variants found in hereditary nonpolyposis colon cancer families that carry missense mutations and where it is unclear whether they predispose to colon cancer. This analysis is made possible by the fact that the human hMSH2 protein is able to complement the MMR defect in the host murine cell line.


Subject(s)
Chromosomes, Human, Pair 2/genetics , Mutation , Proto-Oncogene Proteins/genetics , Alleles , Animals , Base Pair Mismatch , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , DNA Repair/genetics , DNA-Binding Proteins/genetics , Gene Silencing , Humans , Hybrid Cells , Mice , MutS Homolog 2 Protein , Mutation, Missense
14.
Cancer Res ; 61(7): 2813-5, 2001 Apr 01.
Article in English | MEDLINE | ID: mdl-11306449

ABSTRACT

Hereditary nonpolyposis colorectal cancer syndrome is associated with an inherited predisposition to primarily colorectal cancer (CRC) and endometrial cancer (EC); however, the biological basis of the organ involvement remains unknown. As an attempt to explore whether the expression levels of MLH1, MSH2, and MSH6 may play a role, we used immunohistochemistry to study 42 ECs and 35 CRCs from patients carrying the same predisposing mutations. Among MSH2 mutation carriers, MLH1 was expressed in both tumor types, whereas MSH2 and, in many cases, also MSH6, were absent. Remarkably, among MLH1 mutation carriers, 54% of ECs (21 of 39), but none of the CRCs (0 of 32), lacked the MSH2 and/or MSH6 protein in addition to lacking MLH1 protein expression. These results demonstrate a marked difference between hereditary nonpolyposis colorectal cancer-related CRCs and ECs and suggest that the development of the latter tumors is selectively associated with the MSH2/MSH6 protein complex deficiency.


Subject(s)
Colonic Neoplasms/metabolism , Colorectal Neoplasms, Hereditary Nonpolyposis/metabolism , DNA-Binding Proteins/biosynthesis , Endometrial Neoplasms/metabolism , Proto-Oncogene Proteins/biosynthesis , Adaptor Proteins, Signal Transducing , Carrier Proteins , Colonic Neoplasms/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , DNA-Binding Proteins/deficiency , Dimerization , Endometrial Neoplasms/genetics , Female , Germ-Line Mutation , Humans , Immunohistochemistry , MutL Protein Homolog 1 , MutS Homolog 2 Protein , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/deficiency , Nuclear Proteins , Proto-Oncogene Proteins/deficiency
15.
Oncogene ; 1(4): 457-61, 1987.
Article in English | MEDLINE | ID: mdl-3330790

ABSTRACT

Tumor-derived DNA from a non-Hodgkin's (T cell) lymphoma patient, assayed by NIH3T3 transfection followed by inoculation of cells into nude mice, was found to contain an activated N-ras proto-oncogene. The mode of activation was determined by hybridization with N-ras-specific oligonucleotide probes detecting mutations at codons 12, 13 and 61. A transversion in codon 13 (GGT----TGT) resulting in replacement of glycine13 by cysteine13 in ras p21 protein was found. The mutation was detected in DNA from mouse tumors induced by transfected NIH3T3 cells and in DNA from patient tumor lymphoblasts. The patient was heterozygous for this mutation. These data identify the first base of codon 13 as a novel mutation site in ras genes and indicate that cysteine at position 13 of the ras p21 is a transforming substitution.


Subject(s)
Lymphoma, Non-Hodgkin/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogenes , Cell Transformation, Neoplastic/genetics , Codon , DNA, Neoplasm/genetics , Humans , Mutation , Oligodeoxyribonucleotides/genetics , Proto-Oncogene Mas
16.
J Mol Biol ; 276(4): 705-19, 1998 Mar 06.
Article in English | MEDLINE | ID: mdl-9500919

ABSTRACT

We have studied whether spontaneous intrachromosomal recombination is altered in methylation tolerant human cells with a defect in mismatch repair. Somatic recombination was analysed in HeLaMR cells containing the vector pTPSN, which carries two copies of the gene for hygromycin resistance. The hygromycin genes are both inactivated by an inserted HindIII linker but hygromycin-resistant clones can arise by recombination. The spontaneous rate of recombination in a clone of HeLaMR cells containing a single integrated copy of pTPSN (HeLaG1) was 3.1x10(-6)/cell per generation. Two methylation tolerant variants from HeLaG1 cells (clone 12 and clone 15) were isolated by exposure to MNNG. Clone 12 cells exhibited a 16-fold increase in spontaneous mutation rate at the HPRT gene and extensive microsatellite instability at both mono- and dinucleotide repeats. Microsatellite instability limited to mononucleotide repeats was found in clone 15, whereas the mutation rate at HPRT was not significantly affected. A mismatch binding defect in extracts of clone 15 could be complemented by exogenous GTBP but not by purified hMSH2 protein. These data suggest that clone 15 is defective in GTBP. Extracts of clone 12 were unable to correct a single C:T mispair and complementation by extracts of human colorectal carcinoma cells with known deficiencies in mismatch repair indicated a defect in hMutLalpha. Western blotting with antibodies against different human mismatch repair proteins showed that clone 12 cells did not express hPMS2 protein, but expression of hMLH1, hMSH2 and GTBP appeared normal. The spontaneous recombination rate of clone 12 was 19-fold higher than the parental HeLaG1 cells, whereas no increase was observed in clone 15. Analysis of individual recombinants showed that hygromycin resistance arose exclusively by gene conversion. Our data indicate that mismatch correction regulates somatic recombination in human cells.


Subject(s)
Cinnamates , DNA Methylation , DNA Repair/genetics , Recombination, Genetic , Base Sequence , DNA/genetics , DNA/metabolism , DNA Primers/genetics , Drug Resistance, Microbial/genetics , Gene Conversion , Genetic Vectors , HeLa Cells , Humans , Hygromycin B/analogs & derivatives , Hygromycin B/pharmacology , Hypoxanthine Phosphoribosyltransferase/genetics , Methylnitronitrosoguanidine , Microsatellite Repeats , Mutation , Plasmids/genetics , Transfection
17.
Gene ; 65(2): 219-27, 1988 May 30.
Article in English | MEDLINE | ID: mdl-2842233

ABSTRACT

The binding of the transcription factors Sp1 and CTF immediately upstream from the TATA box of the Herpes simplex virus (type 1) thymidine kinase-coding gene (tk) facilitates efficient transcription of this gene in microinjected Xenopus laevis oocytes. To establish whether the presence of methylated CpG dinucleotides within the binding sites of these two factors affects transcription of the tk gene in vivo, we replaced a 33-bp promoter segment, consisting solely of the Sp1 and CTF binding sites, with synthetic oligodeoxynucleotide duplexes containing 5-methylcytosine residues at selected positions. We show that symmetrical methylation (modification of both strands) of any of the four CpGs within this promoter segment resulted in an approximately 20-fold reduction in the specific transcription of the tk gene in Xenopus oocytes, as shown by primer extension analysis of the isolated mRNA. As no other methylated CpG dinucleotides were present within the entire 9.2-kb vector, our results demonstrate that the presence of a single mCpG dinucleotide within the promoter region is sufficient for transcriptional inactivation of the tk gene. The possible mechanisms of this downregulation are discussed.


Subject(s)
Cytidine Monophosphate/metabolism , Cytosine Nucleotides/metabolism , Dinucleoside Phosphates , Genes, Viral , Guanosine/analogs & derivatives , Promoter Regions, Genetic , Simplexvirus/genetics , Transcription, Genetic , Animals , Cytidine Monophosphate/analogs & derivatives , Cytidine Monophosphate/genetics , Female , Genetic Vectors , Guanosine/genetics , Guanosine/metabolism , Methylation , Oocytes , Thymidine Kinase/genetics , Xenopus laevis
18.
Gene ; 41(2-3): 225-31, 1986.
Article in English | MEDLINE | ID: mdl-3519365

ABSTRACT

Mutation of the initiation codon of the dispensible open reading frame, ORF VII, of cauliflower mosaic virus (CaMV) delayed the appearance of disease symptoms, but the mutants reverted with high frequency. This suggests a role of this start codon in viral expression. Oligonucleotide-directed mutagenesis, utilizing a novel, repair-resistant deoxyguanosine analogue, 2'-deoxy-7-deazainosine (dDI), highly improved the yield of mutants.


Subject(s)
DNA Repair , Genes, Viral , Mosaic Viruses/genetics , Mutation , Oligodeoxyribonucleotides/pharmacology , Base Sequence , Brassica , Codon , Escherichia coli/genetics , Mosaic Viruses/drug effects , Oligodeoxyribonucleotides/chemical synthesis , Plasmids
19.
FEBS Lett ; 165(2): 216-22, 1984 Jan 09.
Article in English | MEDLINE | ID: mdl-6319182

ABSTRACT

A 500 MHz 1H-NMR study on a synthetic DNA pentadecamer comprising the specific target site of the cAMP receptor protein in the ara BAD operon is presented. Using pre-steady state NOE measurements, unambiguous assignments of all the imino proton resonances and associated adenine (H2) resonances are obtained. From the NOE data interbase pair interproton distances involving the imino and adenine (H2) protons are determined. It is shown that these distances are very similar to those expected for classical B DNA (RMS difference of 0.5 A), but are significantly different from those expected for classical A DNA (RMS difference of 1.1 A).


Subject(s)
DNA , Magnetic Resonance Spectroscopy , Operon , Receptors, Cyclic AMP/genetics , Adenine , Protons , Temperature
20.
Clin Exp Metastasis ; 17(8): 663-8, 1999.
Article in English | MEDLINE | ID: mdl-10919711

ABSTRACT

Transition from an epithelioid (E) to a round (R) morphotype, in the human colon cancer cell line HCT-8, is associated with loss or truncation of alphaE-catenin and acquisition of invasiveness in organ culture. In E clones, like in parental HCT-8 cells, one allele of the alphaE-catenin gene (CTNNA1) is mutated. HCT-8 cells have also a 'Microsatelite Instability-High' (MSI-H) phenotype presumably due to a mutated hMSH6 gene. Fusion of E type cells doubles the wild type CTNNA1 alleles and prevents the loss of alphaE-catenin. Introduction of an extra chromosome 2, carrying a wild type hMSH6 gene, restores post-replicative mismatch repair and also prevents the frequent inactivation of the remaining wild type CTNNA1 allele.


Subject(s)
Colonic Neoplasms/genetics , Cytoskeletal Proteins/genetics , DNA-Binding Proteins/deficiency , Gene Silencing/physiology , Genes, Tumor Suppressor/genetics , Alleles , Animals , Base Pair Mismatch , Cell Fusion , Chromosomes, Human, Pair 2/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Cytoskeletal Proteins/biosynthesis , DNA Repair , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Mice , Phenotype , Polyploidy , Transfection , Tumor Cells, Cultured , alpha Catenin
SELECTION OF CITATIONS
SEARCH DETAIL