Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Biol Chem ; 382(4): 699-705, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11405234

ABSTRACT

We analysed the methylation patterns of CpG dinucleotides in a bidirectional promoter region (LRS, LMP 1 regulatory sequences) of latent Epstein-Barr virus (EBV) genomes using automated fluorescent genomic sequencing after bisulfite-induced modification of DNA. Transcripts for two latent membrane proteins, LMP 1 (a transforming protein) and LMP 2B, are initiated in this region in opposite directions. We found that B cell lines and a clone expressing LMP 1 carried EBV genomes with unmethylated or hypomethylated LRS, while highly methylated CpG dinucleotides were present at each position or at discrete sites and within hypermethylated regions in LMP 1 negative cells. Comparison of high resolution methylation maps suggests that CpG methylation-mediated direct interference with binding of nuclear factors LBF 2, 3, 7, AML1/LBF1, LBF5 and LBF6 or methylation of CpGs within an E-box sequence (where activators as well as repressors can bind) is not the major mechanism in silencing of the LMP 1 promoter. Although a role for CpG methylation within binding sites of Sp1 and 3, ATF/CRE and a sis-inducible factor (SIF) cannot be excluded, hypermethylation of LRS or regions within LRS in LMP 1 negative cells suggests a role for an indirect mechanism, via methylcytosine binding proteins, in silencing of the LMP 1 promoter.


Subject(s)
DNA, Viral/isolation & purification , Genome, Viral , Herpesvirus 4, Human/genetics , Promoter Regions, Genetic/genetics , Viral Matrix Proteins/genetics , CpG Islands/genetics , DNA Methylation , Humans , Sequence Analysis , Virus Latency
2.
J Biol Chem ; 276(2): 1634-42, 2001 Jan 12.
Article in English | MEDLINE | ID: mdl-11038348

ABSTRACT

p16(INK4a) is frequently altered in human cancer, often through epigenetically mediated transcriptional silencing. However, little is known about the transcriptional regulation of this gene. To learn more about such control, we initiated studies of proteins that bind to the promoter in cancer cells that do, and do not, express the gene. We identify RNA helicase A (RHA) as a protein that binds much better to the p16(INK4a) promoter in the expressing cells. RHA has not previously been characterized to manifest sequence-specific DNA interaction but does so to the sequence 5' CGG ACC GCG TGC GC 3' in the p16(INK4a) promoter. The Drosophila homologue to RHA, maleless (Mle), functions in the fly for 2-fold activation of male X-chromosome genes. In our experimental setting, RHA induces a similar modest up-regulation of the p16(INK4a) promoter that is dependent upon its sequence-specific interaction. Mle colocalizes with hyperacetylated H4Ac16 on the X-chromosome and some autosomal loci. The decreased binding of RHA to p16(INK4a) in our cells, where the gene is transcriptionally inactive, is associated with decreased amounts of RHA that immunoprecipitate with acetylated lysine antibodies. Finally, we show RHA to be a cellular substrate for caspase-3, which decreases its sequence-specific binding to p16(INK4a) by cleavage of the N terminus. Thus, we have identified a new protein interaction with the p16(INK4a) promoter that involves an important protein for transcriptional modulation. This interaction is decreased in cancer cells, where this gene is aberrantly transcriptionally silent.


Subject(s)
Autoantigens/metabolism , Cyclin-Dependent Kinase Inhibitor p16/genetics , Genes, Tumor Suppressor , Promoter Regions, Genetic , RNA Helicases/metabolism , Animals , Autoantigens/genetics , Base Sequence , Binding Sites , Carcinoma, Small Cell , Carcinoma, Squamous Cell , Cell Nucleus/metabolism , DEAD-box RNA Helicases , DNA-Binding Proteins/metabolism , Drosophila/genetics , Gene Expression Regulation , Humans , Lung Neoplasms , Male , Neoplasm Proteins , Protein Biosynthesis , RNA Helicases/genetics , Rabbits , Recombinant Proteins/metabolism , Reticulocytes/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Transcription, Genetic , Transfection , Tumor Cells, Cultured , X Chromosome
3.
Biol Chem ; 381(2): 95-105, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10746740

ABSTRACT

Latent episomal genomes of Epstein-Barr virus, a human gammaherpesvirus, represent a suitable model system for studying replication and methylation of chromosomal DNA in mammals. We analyzed the methylation patterns of CpG dinucleotides in the latent origin of DNA replication of Epstein-Barr virus using automated fluorescent genomic sequencing of bisulfite-modified DNA samples. We observed that the minimal origin of DNA replication was unmethylated in 8 well-characterized human cell lines or clones carrying latent Epstein-Barr virus genomes as well as in a prototype virus producer marmoset cell line. This observation suggests that unmethylated DNA domains can function as initiation sites or zones of DNA replication in human cells. Furthermore, 5' from this unmethylated region we observed focal points of de novo DNA methylation in nonrandom positions in the majority of Burkitt's lymphoma cell lines and clones studied while the corresponding CpG dinucleotides in viral genomes carried by lymphoblastoid cell lines and marmoset cells were completely unmethylated. Clustering of highly methylated CpG dinucleotides suggests that de novo methylation of unmethylated double-stranded episomal viral genomes starts at discrete founder sites in vivo. This is the first comparative high-resolution methylation analysis of a latent viral origin of DNA replication in human cells.


Subject(s)
DNA Methylation , DNA Replication , Genome, Viral , Herpesvirus 4, Human/genetics , Replication Origin/genetics , Virus Replication , Base Sequence , Binding Sites/genetics , Burkitt Lymphoma/genetics , Cell Line , Cytosine/metabolism , DNA Transposable Elements , Electronic Data Processing , Epstein-Barr Virus Nuclear Antigens/genetics , Humans , Lymphocytes , Molecular Sequence Data , Repetitive Sequences, Nucleic Acid , Sequence Analysis, DNA
4.
Nat Genet ; 21(1): 103-7, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9916800

ABSTRACT

Densely methylated DNA associates with transcriptionally repressive chromatin characterized by the presence of underacetylated histones. Recently, these two epigenetic processes have been dynamically linked. The methyl-CpG-binding protein MeCP2 appears to reside in a complex with histone deacetylase activity. MeCP2 can mediate formation of transcriptionally repressive chromatin on methylated promoter templates in vitro, and this process can be reversed by trichostatin A (TSA), a specific inhibitor of histone deacetylase. Little is known, however, about the relative roles of methylation and histone deacetylase activity in the stable inhibition of transcription on densely methylated endogenous promoters, such as those for silenced alleles of imprinted genes, genes on the female inactive X chromosome and tumour-suppressor genes inactivated in cancer cells. We show here that the hypermethylated genes MLH1, TIMP3 (TIMP3), CDKN2B (INK4B, p15) and CDKN2A (INK4, p16) cannot be transcriptionally reactivated with TSA alone in tumour cells in which we have shown that TSA alone can upregulate the expression of non-methylated genes. Following minimal demethylation and slight gene reactivation in the presence of low dose 5-aza-2'deoxycytidine (5Aza-dC), however, TSA treatment results in robust re-expression of each gene. TSA does not contribute to demethylation of the genes, and none of the treatments alter the chromatin structure associated with the hypermethylated promoters. Thus, although DNA methylation and histone deacetylation appear to act as synergistic layers for the silencing of genes in cancer, dense CpG island methylation is dominant for the stable maintenance of a silent state at these loci.


Subject(s)
Cell Cycle Proteins , DNA Methylation , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylase Inhibitors , Hydroxamic Acids/pharmacology , Tumor Suppressor Proteins , Adaptor Proteins, Signal Transducing , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Carrier Proteins/genetics , CpG Islands , Cyclin-Dependent Kinase Inhibitor p15 , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA Modification Methylases/antagonists & inhibitors , Decitabine , Enzyme Inhibitors/pharmacology , Humans , MutL Protein Homolog 1 , Neoplasm Proteins/genetics , Neoplasms/genetics , Nuclear Proteins , Promoter Regions, Genetic , Tissue Inhibitor of Metalloproteinase-3/genetics , Transcriptional Activation/drug effects , Tumor Cells, Cultured
5.
J Biotechnol ; 61(3): 191-8, 1998 May 13.
Article in English | MEDLINE | ID: mdl-9684337

ABSTRACT

To study the expression of the bovine beta-lactoglobulin (BLG) gene we isolated the BLG gene from a genomic library and introduced it into murine germline. Bovine BLG gene including 2.8 kbp of 5' and 1.9 kbp of 3' flanking region was expressed efficiently and mammary gland-specifically in transgenic mice. Expression levels of BLG in milk exceeded 1 mg ml-1 in all four mouse lines analyzed. However, in two mouse lines originating from female founders BLG expression levels varied from less than 0.02 mg ml-1 up to 1 mg ml-1. In both lines originating from male founders all analyzed female mice excreted bovine BLG into their milk at a high and constant level of 1-2 mg ml-1. BLG expression was stable within individual mice in two successive lactations and the amount of BLG in the milk of mice correlated with the level of BLG mRNA in the mammary tissue. Methylation analyses of HpaII sites revealed that transgene copies were on average more methylated in mice which excreted low levels of BLG into their milk. Each mouse line had its own methylation pattern and, in addition, each mouse had more or less identical methylation patterns in mammary gland, brain and kidney DNA. Genomic sequencing of the BLG gene indicated that the promoter region (bases -162 to +391 with respect to the transcription start site) was heavily methylated except for distinct CpG sites that were only partially methylated both in transgenic mice and lactating cattle.


Subject(s)
Lactoglobulins/genetics , Animals , Breast/physiology , Cattle , Cloning, Molecular , DNA Methylation , Female , Gene Dosage , Gene Expression/genetics , Mice , Mice, Transgenic , Milk Proteins/metabolism , Promoter Regions, Genetic/genetics , RNA, Messenger/metabolism , Recombinant Proteins/isolation & purification , Sequence Analysis
6.
Biol Chem ; 379(4-5): 417-22, 1998.
Article in English | MEDLINE | ID: mdl-9628332

ABSTRACT

We analyzed the methylation patterns of CpG dinucleotides in the regulatory region of the latent Epstein-Barrvirus (EBV) promoter BCR2 (also called C promoter, Cp) using automated fluorescent genomic sequencing after bisulfite-induced modification of DNA. BCR2 is one of the alternative promoters for transcripts encoding the growth-transformation-associated nuclear antigens EBNA 1-6 which are expressed in a host cell phenotype dependent manner. Well characterized clones isolated from the Burkitt's lymphoma (BL) line Mutu differing from each other as to their phenotype and EBV latent gene expression were used in the present study. We found that in Mutu BL III clone 99 which is actively using the BCR2 promoter the regulatory sequences are unmethylated with two exceptions (position 10702 and 10799). In contrast, there are 15 methylated cytosines in the same region in Mutu BL I clone 216 where the BCR2 promoter is silent. Cytosines which are potential targets of DNA methyltransferase in the immediate vicinity or within the attachment sites of cellular C promoter binding factors CBF1 and CBF2 remained hypomethylated in Mutu BL I clone 216. This suggests a role for a hypermethylated region (nucleotides 10666 -10865, -639 to -440 bases upstream from the beginning of the TATA box at position 11305) in silencing of the BCR2 promoter in these cells.


Subject(s)
DNA Methylation , DNA, Viral/metabolism , Epstein-Barr Virus Nuclear Antigens/genetics , Promoter Regions, Genetic , Sequence Analysis, DNA , Base Sequence , CpG Islands , Fluorescein , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/physiology , Molecular Sequence Data , Regulatory Sequences, Nucleic Acid , Sequence Analysis, DNA/methods , Virus Latency
7.
Cancer Res ; 58(4): 591-3, 1998 Feb 15.
Article in English | MEDLINE | ID: mdl-9485004

ABSTRACT

Inactivation of p16ink4A and other tumor suppressor genes has been associated with promoter region hypermethylation in neoplasia. However, direct proof for aberrant DNA methylation as an independent event for loss of gene function has been difficult to obtain. We addressed this question in the colon carcinoma cell line HCT116, which contains one allele of p16ink4A with a coding region frameshift mutation and one wild-type allele. Neither allele contains a mutation in the proximal promoter region. The promoter of the wild-type allele, but not the mutant allele, is hypermethylated, and only the mutant allele is expressed. Transcription from the methylated/wild-type allele was restored after cell treatment with the demethylating agent 5-aza-2'-deoxycytidine. Thus, in neoplastic cells, stable allele-specific loss of transcription may arise from aberrant methylation of a nonmutated promoter region, identifying hypermethylation as a direct mechanism for tumor suppressor gene inactivation.


Subject(s)
Colonic Neoplasms/genetics , Cyclin-Dependent Kinases/genetics , DNA Methylation , Gene Expression Regulation, Neoplastic , Genes, p16 , Promoter Regions, Genetic , Alleles , Base Sequence , CpG Islands , Frameshift Mutation , Humans , Tumor Cells, Cultured
8.
J Biol Chem ; 272(35): 22322-9, 1997 Aug 29.
Article in English | MEDLINE | ID: mdl-9268383

ABSTRACT

Promoter region CpG island methylation is associated with tumor suppressor gene silencing in neoplasia. GenBank sequence analyses revealed that a number of CpG islands are juxtaposed to multiple Alu repeats, which have been proposed as "de novo methylation centers." These islands also contain multiple Sp1 elements located upstream and downstream of transcription start, which have been shown to protect CpG islands from methylation. We mapped the methylation patterns of the E-cadherin (E-cad) and von Hippel-Lindau (VHL) tumor suppressor gene CpG island regions in normal and neoplastic cells. Although unmethylated in normal tissue, these islands were embedded between densely methylated flanking regions containing multiple Alu repeats. These methylated flanks were segregated from the unmethylated, island CpG sites by Sp1-rich boundary regions. Finally, in human fibroblasts overexpressing DNA methyltransferase, de novo methylation of the E-cad CpG island initially involved sequences at both ends of the island and the adjacent, flanking regions and progressed with time to encompass the entire CpG island region. Together, these data suggest that boundaries exist at both ends of a CpG island to maintain the unmethylated state in normal tissue and that these boundaries may be progressively overridden, eliciting the de novo methylation associated with tumor suppressor gene silencing in neoplasia.


Subject(s)
CpG Islands , DNA Methylation , DNA, Neoplasm/metabolism , DNA-Cytosine Methylases/metabolism , Base Sequence , Breast/chemistry , Cadherins/genetics , Chromosome Mapping , Female , Genes, Tumor Suppressor , Humans , Molecular Sequence Data , Tumor Cells, Cultured , von Hippel-Lindau Disease/genetics
9.
Int J Cancer ; 70(6): 644-8, 1997 Mar 17.
Article in English | MEDLINE | ID: mdl-9096643

ABSTRACT

Germline mutations of the putative tumor suppressor gene APC are associated in high frequency with the familial adenomatous polyposis, predisposing the patients to colorectal neoplasia. Similarly, sequence analyses have revealed that in more than half of patients with sporadic colorectal carcinoma or adenoma, the APC gene was mutated. By employing genomic sequencing, i.e., base-specific analysis of methylated cytosines, we show here that the promoter region of the APC gene is heavily methylated at CpG sites in patients with colorectal carcinoma in comparison with normal colonic mucosa and premalignant adenomas. Our results suggest that cytosine methylation of the regulatory sequences of the APC gene could be involved in the progression of human colorectal cancer.


Subject(s)
Adenoma/genetics , Colorectal Neoplasms/genetics , Cytosine/metabolism , Genes, APC/genetics , Precancerous Conditions/genetics , Adenoma/metabolism , Colorectal Neoplasms/metabolism , DNA Methylation , Germ-Line Mutation , Humans , Intestinal Mucosa/metabolism , Polymerase Chain Reaction , Precancerous Conditions/metabolism , Promoter Regions, Genetic
10.
Br J Cancer ; 76(9): 1124-30, 1997.
Article in English | MEDLINE | ID: mdl-9365158

ABSTRACT

The short arm of the chromosome 11, known to harbour a number of putative and established tumour-suppressor genes, is frequently hypermethylated in various human neoplasms. We subjected the promoter regions of two genes residing at 11p, namely the tumour-suppressor gene WT1 (Wilms' tumour gene) (11p13) and the calcitonin gene (11p15.5), to methylation analysis in human sporadic colorectal cancer using genomic sequencing. Both genes showed significant hypermethylation of CpG sites within their promoter regions in adenomas and carcinomas compared with normal colonic mucosa. Although the WT1 promoter region was significantly hypermethylated, two CpG sites located in Sp1 motifs were unmethylated in the majority of cases (68-74% of carcinomas). The expression of WT1 gene, as revealed by in situ hybridization, showed no differences between normal colonic mucosa and malignant carcinoma. Together with earlier observations, our present results support the view that the short arm of human chromosome 11 is subjected to widespread regional hypermethylation in various human malignancies.


Subject(s)
Calcitonin/genetics , Colorectal Neoplasms/genetics , DNA-Binding Proteins/genetics , Transcription Factors/genetics , Adenoma/metabolism , Chromosomes, Human, Pair 11 , Colon/metabolism , Colorectal Neoplasms/metabolism , DNA Methylation , DNA-Binding Proteins/metabolism , Genes, Wilms Tumor/genetics , Humans , In Situ Hybridization , Intestinal Mucosa/metabolism , Promoter Regions, Genetic , RNA, Messenger/analysis , Transcription Factors/metabolism , WT1 Proteins
11.
Int J Cancer ; 69(6): 471-4, 1996 Dec 20.
Article in English | MEDLINE | ID: mdl-8980249

ABSTRACT

DNA methylation has been studied intensively during the past years in order to elucidate its role in the regulation of gene expression, gene imprinting and cancer progression. Earlier studies have shown that a general genomic under-methylation is associated with chronic lymphocytic leukemia and metastatic prostate cancer. Site-specific methylation changes, as revealed by the use of methylation-sensitive restriction enzymes, have been reported to occur in the promotor region of the calcitonin gene in chronic myeloid leukemia as it progresses from the chronic phase to blast crisis, in non-Hodgkin's lymphoid neoplasms and in non-lymphocytic leukemia. We have now explored possible methylation changes associated with benign and malignant breast tumors. Two approaches were employed: (i) chemical determination of general genomic methylation status and (ii) base-specific analysis of the methylation changes in the promoter of the calcitonin gene with the aid of genomic sequencing. The results did not reveal any changes of total DNA 5-methylcytosine content in ductal carcinoma of breast in comparison with benign tumors. There was a small, yet significant, increase in 5-methylcytosine content in lobular carcinoma. Genomic sequencing of the promoter region of the calcitonin gene, however, revealed a striking hypermethylation at or around the transcription start site of the gene in ductal carcinomas. In benign tumors and lobular carcinomas, this region was either entirely unmethylated or only slightly methylated. The latter changes may reflect a regional hypermethylation of the short arm of chromosome 11, which harbors, in addition to the calcitonin gene, a number of putative or established tumor-suppressor genes. Our results demonstrate that genomic sequencing in its present form can be used for a reliable and precise DNA methylation analysis of primary human tumors.


Subject(s)
Breast Neoplasms/genetics , Calcitonin/genetics , DNA Methylation , DNA, Neoplasm/chemistry , Regulatory Sequences, Nucleic Acid/genetics , 5-Methylcytosine , Breast Neoplasms/chemistry , Carcinoma, Ductal, Breast/chemistry , Carcinoma, Ductal, Breast/genetics , Carcinoma, Lobular/chemistry , Carcinoma, Lobular/genetics , Chromatography, High Pressure Liquid/methods , Cytosine/analogs & derivatives , Cytosine/analysis , Female , Fibroadenoma/chemistry , Fibroadenoma/genetics , Gene Expression Regulation, Neoplastic , Humans , Sequence Analysis, DNA/methods
12.
Proc Natl Acad Sci U S A ; 93(18): 9821-6, 1996 Sep 03.
Article in English | MEDLINE | ID: mdl-8790415

ABSTRACT

Precise mapping of DNA methylation patterns in CpG islands has become essential for understanding diverse biological processes such as the regulation of imprinted genes, X chromosome inactivation, and tumor suppressor gene silencing in human cancer. We describe a new method, MSP (methylation-specific PCR), which can rapidly assess the methylation status of virtually any group of CpG sites within a CpG island, independent of the use of methylation-sensitive restriction enzymes. This assay entails initial modification of DNA by sodium bisulfite, converting all unmethylated, but not methylated, cytosines to uracil, and subsequent amplification with primers specific for methylated versus unmethylated DNA. MSP requires only small quantities of DNA, is sensitive to 0.1% methylated alleles of a given CpG island locus, and can be performed on DNA extracted from paraffin-embedded samples. MSP eliminates the false positive results inherent to previous PCR-based approaches which relied on differential restriction enzyme cleavage to distinguish methylated from unmethylated DNA. In this study, we demonstrate the use of MSP to identify promoter region hypermethylation changes associated with transcriptional inactivation in four important tumor suppressor genes (p16, p15, E-cadherin, and von Hippel-Lindau) in human cancer.


Subject(s)
Cell Cycle Proteins , CpG Islands , Ligases , Polymerase Chain Reaction/methods , Tumor Suppressor Proteins , Ubiquitin-Protein Ligases , Antisense Elements (Genetics)/chemistry , Base Sequence , Cadherins/genetics , Carrier Proteins/genetics , Cell Line , Cyclin-Dependent Kinase Inhibitor p15 , Cyclin-Dependent Kinase Inhibitor p16 , DNA Primers/chemical synthesis , Genes, Tumor Suppressor , Humans , Methylation , Molecular Sequence Data , Proteins/genetics , Restriction Mapping , Von Hippel-Lindau Tumor Suppressor Protein
13.
Carcinogenesis ; 17(5): 1179-81, 1996 May.
Article in English | MEDLINE | ID: mdl-8640933

ABSTRACT

The kinds of mutations induced by 3-chloro-4-(dichloromethyl)-5-hydroxy-2(5H)-furanone (MX) in the protein coding region of the hprt gene of Chinese hamster ovary (CHO) cells were determined by direct sequencing of polymerase chain reaction (PCR)-amplified cDNA. Primary mutations were found in 15 of 19 of the mutants: 11 were G:C-->T:A transversions, two were A:T-->T:A transversions and two were deletions of single G:C base pairs (-1 frameshifts). The remaining four mutants had large alterations in the cDNA that were explained by mRNA splicing errors. A group of control mutants had more diverse hprt cDNA alterations than MX-induced mutants. Transversions yielding an A:T base pair were the predominant type of MX-induced mutations, in agreement with previous findings in bacteria. This specificity may be explained by the 'A rule', that DNA polymerases preferentially insert adenine nucleotides opposite non-instructional lesions.


Subject(s)
Furans/toxicity , Hypoxanthine Phosphoribosyltransferase/genetics , Mutagens/toxicity , Mutation , Animals , Base Sequence , CHO Cells , Cricetinae , Molecular Sequence Data
14.
DNA Cell Biol ; 14(10): 841-7, 1995 Oct.
Article in English | MEDLINE | ID: mdl-7546290

ABSTRACT

We have isolated and sequenced cDNA clones that encode human spermine synthase (EC 2.5.1.22). The total length of the sequenced cDNA was 1,612 nucleotides, containing an open reading frame encoding a polypeptide chain of 368 amino acids. All of the previously sequenced peptide fragments of human and bovine spermine synthase proteins could be located within the coding region derived from the cDNA. An unusual sequence of AATTAA apparently signaled the initiation of polyadenylation. Sequence comparisons between human spermine synthase and spermidine synthases from bacterial and mammalian sources revealed a nearly complete lack of similarity between the primary structures of these two enzymes catalyzing almost identical reactions. A modest similarity found was restricted to a relatively short peptide domain apparently involved in the binding of decarboxylated S-adenosylmethionine, the common substrate for both enzymes. The apparent lack of an overall similarity may indicate that spermine synthase, the enzyme found only in eukaryotes, and spermidine synthase with more universal distribution, although functionally closely related, have evolved separately.


Subject(s)
DNA, Complementary/genetics , Genes , Spermine Synthase/genetics , Amino Acid Sequence , Animals , Base Sequence , Brain/enzymology , Cattle , Cloning, Molecular , Humans , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Polymerase Chain Reaction , Sequence Alignment , Sequence Homology, Amino Acid , Species Specificity , Spermidine Synthase/genetics , Spermine/biosynthesis , Substrate Specificity
15.
Biochem J ; 310 ( Pt 1): 299-303, 1995 Aug 15.
Article in English | MEDLINE | ID: mdl-7646459

ABSTRACT

(1) Human myeloma cell line Sultan, resistant to 20 mM difluoro-methylornithine (DFMO) owing to ornithine decarboxylase (ODC) gene amplification, was grown in the absence of DFMO for a period of 10 months. The gene copy number and methylation status of the ODC gene were monitored after withdrawal of DFMO. Moreover, levels of ODC mRNA, immunoreactive ODC protein, ODC activity and polyamine levels were recorded recurrently during the course of the study. (2) The results revealed that ODC gene copy number started to decrease after 4 weeks growth without DFMO, to a final level of less than 30% of the original gene dosage. The methylation status of the ODC gene, however, remained almost unaltered, displaying only a modest increase in methylation after 10 months without DFMO. The amount of ODC message dropped very rapidly to 75% of the original value, then started to decrease in a gene copy-number-dependent manner. The amount of ODC protein closely followed the levels of mRNA during the study, whereas the ODC activity, after a transient increase during the first week, decreased to half of the original level after 4 weeks. Between 6 and 16 weeks ODC activity stabilized to a fifth of the original value and no more changes were detected during the subsequent period of observation. (3) Due to the grossly elevated ODC enzyme activity, levels of putrescine and spermidine first peaked and then stabilized at 6 weeks after DFMO withdrawal. The final spermidine level was comparable with that of the parental Sultan cell line with only one copy of active ODC gene. However, putrescine content was strikingly elevated, being stabilized to a level that was 20 times higher than in parental cells. Spermine concentration was practically unchanged during the study. (4) According to the results obtained in this study, the abnormal level of ODC expression in human myeloma cells is suppressed partially at the level of transcription or post-transcriptionally, but it is not due to increased methylation of the gene. The major regulatory mechanism to compensate for a highly elevated ODC expression was modulation of the enzyme activity. After 10 months without DFMO, the cells still displayed about 20 times higher ODC activity and putrescine concentration than the myeloma cell line with a single copy of the ODC gene. They did not, however, show any signs of growth retardation or other features different from the parental cells.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Gene Amplification , Multiple Myeloma/enzymology , Ornithine Decarboxylase/genetics , Animals , Eflornithine/pharmacology , Humans , Mice , Mice, Transgenic , Multiple Myeloma/genetics , Ornithine Decarboxylase/metabolism , Polyamines/metabolism , RNA, Messenger/metabolism , Transcription, Genetic , Tumor Cells, Cultured
16.
Gene ; 157(1-2): 325-9, 1995 May 19.
Article in English | MEDLINE | ID: mdl-7607521

ABSTRACT

The DNA methylation pattern of the promoter (pNF-L) region of the rat light-neurofilament-encoding gene (NF-L), a neuron-specific gene, was assessed in NF-L expressing and non-expressing cell lines and tissues by genomic sequencing using PCR amplification of bisulfite-modified DNA. We analysed twenty-five potential CpG methylation sites between nucleotide (nt) positions -311 and +103 of pNF-L, containing Sp1- and AP-2-binding sites, a CGCCCCCGC box and a cAMP-responsive element. Six out of 25 possible CpG methylation sites are within these elements. The pNF-L promoter was unmethylated in NF-L-expressing rat brain, as well as in liver not expressing NF-L. In NF-L-expressing PC12 cells, the promoter was unmethylated, whereas in non-expressing glioma C6 cells intensive methylation occurred. A cluster of methylated CpG dinucleotides spanned the region from nt -176 to -67 bp. Thus, methylation of this promoter region could play a role in silencing NF-L in the glioma cell line in vitro, but not in liver tissue in vivo. In a non-CpG sequence, in the CpApG trinucleotide at nt position -114, cytosine was found to be partially methylated. It is thus possible to describe the methylation state of each cytosine present in the area of genomic DNA of interest.


Subject(s)
DNA/metabolism , Gene Expression , Neurofilament Proteins/biosynthesis , Neurofilament Proteins/genetics , Promoter Regions, Genetic , Animals , Base Sequence , Binding Sites , Cell Line , DNA/chemistry , DNA/genetics , Methylation , Molecular Sequence Data , RNA, Messenger/biosynthesis , Rats , Sequence Homology, Nucleic Acid , Sulfites , Transcription Factors/metabolism , Transcription, Genetic
17.
Mol Cell Biol ; 14(10): 6983-95, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7935415

ABSTRACT

The diazepam-binding inhibitor (DBI; also called acyl coenzyme A-binding protein or endozepine) is a 10-kDa polypeptide found in organisms ranging from yeasts to mammals. It has been shown that DBI and its processing products are involved in various specific biological processes such as GABAA/benzodiazepine receptor modulation, acyl coenzyme A metabolism, steroidogenesis, and insulin secretion. We have cloned and sequenced the Drosophila melanogaster gene and cDNA encoding DBI. The Drosophila DBI gene encodes a protein of 86 amino acids that shows 51 to 56% identity with previously known DBI proteins. The gene is composed of one noncoding 5' and two coding exons and is localized on the chromosomal map at position 65E. Several transcription initiation sites were detected by RNase protection and primer extension experiments. Computer analysis of the promoter region revealed features typical of housekeeping genes, such as the lack of TATA and CCAAT elements. However, in its low GC content and lack of a CpG island, the region resembles promoters of tissue-specific genes. Northern (RNA) analysis revealed that the expression of the DBI gene occurred from the larval stage onwards throughout the adult stage. In adult flies, DBI mRNA and immunoreactivity were detected in the cardia, part of the Malpighian tubules, the fat body, and gametes of both sexes. Developmentally regulated expression, disappearing during metamorphosis, was detected in the larval and pupal brains. No expression was detected in the adult nervous system. On the basis of the expression of DBI in some but not all tissues with high energy consumption, we propose that in D. melanogaster, DBI is involved in energy metabolism in a manner that depends on the substrate used for energy production.


Subject(s)
Carrier Proteins/genetics , Drosophila melanogaster/genetics , Genes, Insect/genetics , Amino Acid Sequence , Animals , Base Sequence , Carrier Proteins/isolation & purification , Chromosome Mapping , Cloning, Molecular , DNA, Complementary/genetics , Diazepam Binding Inhibitor , Drosophila melanogaster/embryology , Drosophila melanogaster/growth & development , Genomic Library , Immunohistochemistry , In Situ Hybridization , Molecular Sequence Data , Promoter Regions, Genetic/genetics , RNA, Messenger/isolation & purification , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Tissue Distribution , Transcription, Genetic
18.
Biotechnology (N Y) ; 12(6): 606-8, 1994 Jun.
Article in English | MEDLINE | ID: mdl-7764950

ABSTRACT

We have generated a transgenic calf from in vitro produced bovine embryos which had undergone transgene analysis and sexing prior to the embryo transfer. Bovine oocytes were isolated from slaughter-house-derived ovaries, matured and fertilized in vitro and subsequently microinjected with a dam-methylated gene construct consisting of genomic sequences encoding human erythropoietin and governed by bovine alpha S1-casein regulatory sequences. After 6 to 7 days in culture, the embryos were biopsied and while the embryo remained in culture, the biopsy was subjected to transgene analysis and sexing. The transgene analysis was accomplished with a combined treatment of the embryo lysates with DpnI restriction endonuclease and Bal31 exonuclease followed by polymerase chain reaction (PCR). The transgene analysis was based on the fact that DpnI only cleaves its recognition sequence if the adenine in the sequence is methylated. Pregnancy was induced by the transfer of three viable female embryos with a distinct transgene signal to a hormonally synchronized heifer recipient. Amniotic fluid analysis performed two months after the embryo transfer confirmed the presence of the transgene. The calf born was found to be transgenic by PCR analysis from blood, ear and fetal membranes. The presence of the transgene was also confirmed by Southern blotting.


Subject(s)
Animals, Genetically Modified , Embryo, Mammalian/physiology , Fertilization in Vitro , Sex Determination Analysis/methods , Animals , Base Sequence , Blotting, Southern , Caseins/genetics , Cattle , DNA Primers , Deoxyribonucleases, Type II Site-Specific , Endodeoxyribonucleases , Erythropoietin/biosynthesis , Erythropoietin/genetics , Female , Humans , Male , Mice , Mice, Transgenic , Molecular Sequence Data , Oocytes/cytology , Oocytes/physiology , Polymerase Chain Reaction/methods , Pregnancy , Regulatory Sequences, Nucleic Acid , Restriction Mapping
19.
DNA Seq ; 4(5): 343-6, 1994.
Article in English | MEDLINE | ID: mdl-7803821

ABSTRACT

The nucleotide sequence of mouse cDNA for spermidine synthase appeared to contain 75 nucleotides of 5' untranslated region, an open reading frame of 909 nucleotides and 297 nucleotides of 3' untranslated region. The open reading frame encoded a polypeptide of 302 amino acids, displaying 95% similarity to human and 33% similarity to E. coli spermidine synthase. The 3' flanking region contained an unusual polyadenylation signal AATACA.


Subject(s)
Spermidine Synthase/genetics , Amino Acid Sequence , Animals , Base Sequence , DNA, Complementary , Humans , Mice , Molecular Sequence Data , Sequence Homology, Amino Acid
20.
DNA Seq ; 5(1): 1-8, 1994.
Article in English | MEDLINE | ID: mdl-7894054

ABSTRACT

A genomic sequencing method for an automated DNA sequencer was developed. The method described here is an improved version of the previously published protocol, which utilizes bisulfite-induced modification of genomic DNA. In our method, the modified DNA is purified without a time-consuming dialysis, and the subsequent 2-step DNA amplification is carried out with one biotinylated primer in order to separate and isolate the strands of the product with the aid of streptavidin-coated magnetic beads. The strands are then sequenced with fluorescent primers and automated DNA sequencer. This provides means to determine reliably the methylation status of cytosines as well as the degree of methylation in a given CpG, site of the target sequence. The method was successfully applied to analyze the promoter region and the 11th exon of the human ornithine decarboxylase ODC gene in various human myeloma cell lines. The study revealed a totally unmethylated promoter region in every cell line studied, whereas the protein coding region appeared to be extensively methylated, although a dexamethasone resistant cell line displayed demethylation in certain CpG sequences. Also, a previously unknown ODC allele was detected.


Subject(s)
Ornithine Decarboxylase/genetics , Sequence Analysis, DNA/methods , Automation , Base Sequence , Cell Line , DNA Primers , Fluorescent Dyes , Humans , Methylation , Molecular Sequence Data
SELECTION OF CITATIONS
SEARCH DETAIL
...