Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Oncogene ; 31(27): 3244-53, 2012 Jul 05.
Article in English | MEDLINE | ID: mdl-22081068

ABSTRACT

Many tumors exhibit elevated chromosome mis-segregation termed chromosome instability (CIN), which is likely to be a potent driver of tumor progression and drug resistance. Causes of CIN are poorly understood but probably include prior genome tetraploidization, centrosome amplification and mitotic checkpoint defects. This study identifies epigenetic alteration of the centromere as a potential contributor to the CIN phenotype. The centromere controls chromosome segregation and consists of higher-order repeat (HOR) alpha-satellite DNA packaged into two chromatin domains: the kinetochore, harboring the centromere-specific H3 variant centromere protein A (CENP-A), and the pericentromeric heterochromatin, considered important for cohesion. Perturbation of centromeric chromatin in model systems causes CIN. As cancer cells exhibit widespread chromatin changes, we hypothesized that pericentromeric chromatin structure could also be affected, contributing to CIN. Cytological and chromatin immunoprecipitation and PCR (ChIP-PCR)-based analyses of HT1080 cancer cells showed that only one of the two HORs on chromosomes 5 and 7 incorporate CENP-A, an organization conserved in all normal and cancer-derived cells examined. Contrastingly, the heterochromatin marker H3K9me3 (trimethylation of H3 lysine 9) mapped to all four HORs and ChIP-PCR showed an altered pattern of H3K9me3 in cancer cell lines and breast tumors, consistent with a reduction on the kinetochore-forming HORs. The JMJD2B demethylase is overexpressed in breast tumors with a CIN phenotype, and overexpression of exogenous JMJD2B in cultured breast epithelial cells caused loss of centromere-associated H3K9me3 and increased CIN. These findings suggest that impaired maintenance of pericentromeric heterochromatin may contribute to CIN in cancer and be a novel therapeutic target.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Centromere/genetics , Centromere/metabolism , Chromosomal Instability , Heterochromatin/metabolism , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/pathology , Cell Line, Tumor , Chromosomes, Human, Pair 5/genetics , Female , Histones/metabolism , Humans , Jumonji Domain-Containing Histone Demethylases/metabolism , Kinetochores/metabolism , Neoplasm Invasiveness
2.
Gene Ther ; 12(21): 1559-72, 2005 Nov.
Article in English | MEDLINE | ID: mdl-15973438

ABSTRACT

Efficient transfer of chromosome-based vectors into mammalian cells is difficult, mostly due to their large size. Using a genetically engineered invasive Escherichia coli vector, alpha satellite DNA cloned in P1-based artificial chromosome was stably delivered into the HT1080 cell line and efficiently generated human artificial chromosomes de novo. Similarly, a large genomic cystic fibrosis transmembrane conductance regulator (CFTR) construct of 160 kb containing a portion of the CFTR gene was stably propagated in the bacterial vector and transferred into HT1080 cells where it was transcribed, and correctly spliced, indicating transfer of an intact and functional locus of at least 80 kb. These results demonstrate that bacteria allow the cloning, propagation and transfer of large intact and functional genomic DNA fragments and their subsequent direct delivery into cells for functional analysis. Such an approach opens new perspectives for gene therapy.


Subject(s)
Cell Line, Tumor/microbiology , DNA, Recombinant/metabolism , Escherichia coli/genetics , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Genome, Bacterial , Cell Line, Tumor/metabolism , Chromosomes, Artificial, Bacterial , Chromosomes, Artificial, Human , Clone Cells , Electroporation , Flow Cytometry , Gene Expression , Humans , In Situ Hybridization, Fluorescence , Lung Neoplasms , Recombination, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Sarcoma
3.
Cytogenet Genome Res ; 107(1-2): 9-13, 2004.
Article in English | MEDLINE | ID: mdl-15305049

ABSTRACT

Human artificial chromosomes (HACs) were generated by transfer of telomerized PAC constructs containing alpha satellite DNA of various human chromosomes. To monitor which cells took up constructs and subsequently formed stable clones under blasticidin S (BS) selection, a CMV/EGFP expression cassette was inserted into a HAC construct based on chromosome 5 alpha satellite DNA (142 kb). Lipofection into HT1080 cells resulted in a small proportion of cells exhibiting bright green fluorescence on day 1. Areas containing such early green cells were marked, and plates monitored over 2 weeks. In only one out of 41 marked areas, a viable clone developed. In the remaining 40 areas, the green cells ceased division at 1-8 cells. In contrast, outside the marked areas, 16 stable clones formed which did not exhibit green fluorescence during the first cell divisions, but all cells of each became green around day 4-6. Fluorescence in situ hybridization (FISH) analysis of isolated clonal lines demonstrated low copy HAC formation without integration. We conclude that transient expression of an EGFP marker on HAC DNA is not a suitable means for the identification of the proportion of transfected cells which are capable of forming viable clones. One explanation could be that the high copy number required to consistently detect transient EGFP expression (Schindelhauer and Laner, 2002) impairs viability and clone formation.


Subject(s)
Chromosomes, Artificial, Human/genetics , Cytomegalovirus/genetics , Green Fluorescent Proteins/genetics , Cell Line , Chromosomes, Artificial, P1 Bacteriophage/genetics , Green Fluorescent Proteins/biosynthesis , Humans , Telomere/genetics , Transfection/methods
4.
Gene Ther ; 9(11): 727-30, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12032697

ABSTRACT

For the development of human artificial chromosomes (HACs) as a gene transfer vehicle we need to assess the efficiency of de novo chromosome formation depending on the type and the copy number of transferred DNA constructs. In order to check transient EGFP expression as a reporter to immediately detect presence of transfected DNA, we microinjected approximately 1 to 10(5) copies of pEGFP-N1 plasmid into the nucleus of various cell types. Whether using primary, immortalized, or tumor cells, at least 10(3)-10(4) copies were required to generate a visible green signal in the majority of the 50-90% of cells surviving injection. Generally, the cells showed relatively constant, copy number-dependent signals. 10(3) copies resulted in faint and 10(5) in bright fluorescence under the microscope. In addition, the different copy number groups contained a small fraction of cells showing much stronger fluorescence, indicating activation or lack of suppression which facilitates detection of as few as 10(2) transferred copies in rare instances. Thus, transient expression from single copies is not sufficient to reliably detect presence of DNA in the nucleus. The result is relevant for the development of low copy HAC transfer protocols.


Subject(s)
Chromosomes, Artificial, Human , Gene Transfer Techniques , Luminescent Proteins/genetics , Gene Expression , Green Fluorescent Proteins , Humans , Microinjections , Transfection/methods
5.
EMBO Rep ; 2(10): 910-4, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11571265

ABSTRACT

We have investigated the potential of PAC-based vectors as a route to the incorporation of a gene in a mammalian artificial chromosome (MAC). Previously we demonstrated that a PAC (PAC7c5) containing alpha-satellite DNA generated mitotically stable MACs in human cells. To determine whether a functional HPRT gene could be assembled in a MAC, PAC7c5 was co-transfected with a second PAC containing a 140 kb human HPRT gene into HPRT-deficient HT1080 cells. Lines were isolated containing a MAC hybridizing with both alpha-satellite and HPRT probes. The MACs segregated efficiently, associated with kinetochore proteins and stably expressed HPRT message after 60 days without selection. Complementation of the parental HPRT deficiency was confirmed phenotypically by growth on HAT selection. These results suggest that MACs could be further developed for delivering a range of genomic copies of genes into cells and that stable transgene expression can be achieved.


Subject(s)
Chromosomes, Artificial , Gene Expression , Genetic Techniques , Hypoxanthine Phosphoribosyltransferase/genetics , Blotting, Northern , Cell Division , Cell Line , DNA, Complementary/metabolism , Humans , In Situ Hybridization, Fluorescence , Microsatellite Repeats , Microscopy, Fluorescence , Mitosis , Phenotype , Time Factors , Transfection , Transgenes
6.
Hum Mol Genet ; 9(11): 1623-31, 2000 Jul 01.
Article in English | MEDLINE | ID: mdl-10861289

ABSTRACT

Mammalian artificial chromosomes (MACs) form in HT1080 cells after transfecting linear yeast artificial chromosome constructs minimally containing competent alphoid arrays, a selectable marker and terminal human telomere repeats. Restrictions on the structure of input DNA in MAC formation were investigated by transfecting circular or linear alphoid constructs with or without human telomere arrays and by varying the position and orientation of the telomere arrays on input linear constructs. Circular input DNA efficiently produced MACs. Absence of telomere arrays from circular input molecules did not significantly alter MAC formation rates. Linear constructs capped with telomere arrays generated MACs effectively, but a severe reduction in MAC formation was observed from linear constructs lacking telomere arrays. Human telomere arrays positioned 1-5 kb from linear construct ends and in either orientation were able to promote MAC formation with similar efficiencies. Both circular and linear input constructs generated artificial chromosomes that efficiently segregated in the absence of selection. Telomeres were not detected on the MACs, regardless of the inclusion of telomere arrays on input DNA, suggesting that circular chromosomes were formed. We found no evidence for acquisition of host cell DNA, which is consistent with de novo chromosome assembly.


Subject(s)
Chromosomes, Human/genetics , DNA, Satellite/genetics , Telomere/genetics , Bacteriophage P1/genetics , Chromosomes, Artificial, Yeast , DNA, Circular/genetics , Humans , In Situ Hybridization, Fluorescence , Repetitive Sequences, Nucleic Acid , Tumor Cells, Cultured
7.
Bioessays ; 21(1): 76-83, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10070257

ABSTRACT

Two reports have shown that mammalian artificial chromosomes (MAC) can be constructed from cloned human centromere DNA and telomere repeats, proving the principle that chromosomes can form from naked DNA molecules transfected into human cells. The MACs were mitotically stable, low copy number and bound antibodies associated with active centromeres. As a step toward second-generation MACs, yeast and bacterial cloning systems will have to be adapted to achieve large MAC constructs having a centromere, two telomeres, and genomic copies of mammalian genes. Available construction techniques are discussed along with a new P1 artificial chromosome (PAC)-derived telomere vector (pTAT) that can be joined to other PACs in vitro, avoiding a cloning step during which large repetitive arrays often rearrange. The PAC system can be used as a route to further define the optimal DNA elements required for efficient MAC formation, to investigate the expression of genes on MACs, and possibly to develop efficient MAC-delivery protocols.


Subject(s)
Centromere/genetics , Chromosomes , DNA/genetics , Genetic Engineering , Animals , Chromosomes, Artificial, Yeast , Cloning, Molecular , Humans
8.
Hum Mol Genet ; 7(12): 1873-86, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9811930

ABSTRACT

Glial cell line-derived neurotrophic factor (GDNF), a distant member of the TGF-beta superfamily, is a survival factor for various neurons, making it a potential therapeutic agent for neurodegenerative disorders. Here we present the genomic structure and characterization of the promoter of the human GDNF (hGDNF) gene. It contains three exons coding for a cDNA of 4.6 kb including large 5'- and 3'-untranslated regions (UTRs). The 3'-UTR contains a polymorphic AGG repeat that appears not to be expanded in patients suffering from different neurodegenerative disorders. RT-PCR results in at least three different hGDNF transcripts including one that lacks exon 2. Transient expression experiments reveal that exon 2 is essential for proper cellular processing to yield a secreted form of hGDNF, whereas expression of exon 3 alone is sufficient to code for a mature form of hGDNF retained within the cell. Our data show that the hGDNF gene is driven by a TATA-containing promoter preceding exon 1. A second promoter element has been mapped to a region 5' of exon 2. Both promoters are in close proximity to CpG islands covering exons 1 and 2. Using luciferase as a reporter gene, the TATA-containing hGDNF promoter facilitates a 20- to 40-fold increase in transcription when compared with a corresponding promoterless construct, whereas the second promoter confers only weak activity. Furthermore, fibroblast growth factor 2, tetradecanoyl 12-phorbol acetate, an inflammatory agent, and cAMP increase promoter activity, suggesting that GDNF transcriptional regulation is a target of exogenous signals.


Subject(s)
Alternative Splicing/genetics , Exons/genetics , Genes/genetics , Nerve Growth Factors , Nerve Tissue Proteins/genetics , Trinucleotide Repeats/genetics , Amino Acid Sequence , Animals , Bacteriophage P1/genetics , Base Sequence , Carcinogens/pharmacology , Cell Line , Cyclic AMP/pharmacology , DNA/chemistry , DNA/genetics , DNA, Complementary/genetics , DNA, Recombinant , Databases, Factual , Eukaryotic Cells/cytology , Eukaryotic Cells/drug effects , Eukaryotic Cells/metabolism , Fibroblast Growth Factor 2/pharmacology , Gene Expression , Gene Expression Regulation/drug effects , Gene Library , Genetic Vectors , Glial Cell Line-Derived Neurotrophic Factor , Humans , Introns/genetics , Mice , Molecular Sequence Data , Neurodegenerative Diseases/genetics , Polymorphism, Genetic , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/physiology , Recombinant Fusion Proteins/genetics , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Tetradecanoylphorbol Acetate/pharmacology , Transcription, Genetic/genetics , Tumor Cells, Cultured
9.
Eur J Med Res ; 3(3): 149-56, 1998 Mar 23.
Article in English | MEDLINE | ID: mdl-9502755

ABSTRACT

Numerous gene mutations associated with hereditary disorders have been identified. In cystic fibrosis the hereditary defect is attributed to mutations in one single gene, the gene coding for the cystic fibrosis transmembrane conductance regulator (CFTR). Conventional therapies of CF have dramatically increased the life expectancy of afflicted individuals. However, the ultimate incurability of this disease calls for novel and better therapeutic strategies. As cystic fibrosis is believed to be caused by mutations in one single gene, it has appeared to be the ideal candidate for one of the most tempting approaches in clinical therapy, namely gene therapy. Laboratory protocols for the introduction of genes into various tissues have been developed and applied over the last 15 years. The ease of gene transfer under laboratory conditions gave rise to the hope that rapid advances in gene transfer protocols under clinical settings could be achieved as well. 20 clinical trials of gene therapy for cystic fibrosis have been initiated using viral and non-viral vectors for gene transfer (Marcel and David Grausz 1997). The outcome of the CF gene therapy studies as well as of those for other diseases have clearly demonstrated that gene transfer and gene therapy in humans is a much more complex and challenging task than originally thought. Still, the encouraging results achieved in animal models and the rapid progress in vector technology justify the hope that the novel genetic therapies will be applied successfully to the benefit of patients suffering from cystic fibrosis.


Subject(s)
Cystic Fibrosis/therapy , Genetic Therapy , Gene Expression Regulation , Genetic Vectors , Humans
10.
Nucleic Acids Res ; 25(11): 2241-3, 1997 Jun 01.
Article in English | MEDLINE | ID: mdl-9153331

ABSTRACT

In an attempt to combine a cloned genomic copy of a selectable gene with different cloned centromeric sequences to develop mammalian artificial chromosomes (MAC) we used site specific recombination mediated by purified Cre recombinase acting on the loxP sequence in PAC vector DNA. A new method was required to purify highly concentrated, virtually 100% intact PAC DNA which could be stored for a long period. Here we show the efficient linking of linearized PACs containing alpha satellite DNA from chromosomes X and 17 with sizes of 125 and 140 kb, respectively, to a 95 kb restriction fragment derived from a 175 kb PAC containing the intact human HPRT gene locus.


Subject(s)
DNA/metabolism , Genetic Techniques , Hypoxanthine Phosphoribosyltransferase/genetics , Recombination, Genetic , Viral Proteins , Bacteriophage P1 , Chromosomes , Chromosomes, Bacterial , Chromosomes, Human, Pair 17 , DNA, Circular/metabolism , DNA, Satellite/metabolism , DNA, Viral/chemistry , Humans , In Vitro Techniques , Integrases/metabolism , X Chromosome
11.
Genome Res ; 6(11): 1056-69, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8938429

ABSTRACT

Most of the yeast artificial chromosomes (YACs) isolated from the Xp11.23-22 region have shown instability and chimerism and are not a reliable resource for determining physical distances. We therefore constructed a long-range pulsed-field gel electrophoresis map that encompasses approximately 3.5 Mb of genomic DNA between the loci TIMP and DXS146 including a CpG-rich region around the WASP and TFE-3 gene loci. A combined YAC-cosmid contig was constructed along the genomic map and was used for fine-mapping of 15 polymorphic microsatellites and 30 expressed sequence tags (ESTs) or sequence transcribed sites (STSs), revealing the following order: tel-(SYN-TIMP)-(DXS426-ELK1)-ZNF(CA) n-L1-DXS1367-ZNF81-ZNF21-DXS6616- (HB3-OATL1pseudogenes-DXS6950)-DXS6949-DXS694 1-DXS7464E(MG61)-GW1E(EBP)- DXS7927E(MG81)-RBM- DXS722-DXS7467E(MG21)-DXS1011E-WASP-DXS6940++ +-DXS7466E(MG44)-GF1- DXS226-DXS1126-DXS1240-HB1- DXS7469E-(DXS6665-DXS1470)-TFE3-DXS7468E-+ ++SYP-DXS1208-HB2E-DXS573-DXS1331- DXS6666-DXS1039-DXS 1426-DXS1416-DXS7647-DXS8222-DXS6850-DXS255++ +-CIC-5-DXS146-cen. A sequence-ready map was constructed for an 1100-kb gene-rich interval flanked by the markers HB3 and DXS1039, from which six novel ESTs/STSs were isolated, thus increasing the number of markers used in this interval to thirty. This precise ordering is a prerequisite for the construction of a transcription map of this region that contains numerous disease loci, including those for several forms of retinal degeneration and mental retardation. In addition, the map provides the base to delineate the corresponding syntenic region in the mouse, where the mutants scurfy and tattered are localized.


Subject(s)
Chromosome Mapping , X Chromosome/genetics , Amino Acid Sequence , Animals , Base Sequence , Chromosomes, Artificial, Yeast , Cosmids/genetics , DNA Probes/genetics , Electrophoresis, Gel, Pulsed-Field , Genetic Markers/genetics , Humans , Mice , Microsatellite Repeats , Molecular Sequence Data , Sequence Analysis , Zinc Fingers/genetics
12.
Hum Genet ; 98(1): 68-76, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8682510

ABSTRACT

The Wiskott-Aldrich syndrome protein (WASP) gene was found to be mutated in patients presenting with WAS and in patients showing X-linked thrombocytopenia. Mutation analysis in 19 families of German, Swiss and Turkish descent by single-strand conformation polymorphism and sequencing resulted in the detection of seven novel and 10 known mutations. A striking clustering of missense mutations in the first four exons contrasted with a random distribution of nonsense mutations. More than 85% of all known missense mutations were localized in the amino-terminal stretch of the WASP gene product; this region contained a mutational hot spot at codon 86. No genotype-phenotype correlation emerged after a comparison of the identified mutations with the resulting clinical picture for a classical WAS phenotype. A substitution at codon 86 resulted in an extremely variable expression of the disease in a large Swiss family. An extended homology search revealed a distant relationship of this stretch to the vasodilator-stimulated phosphoprotein (VASP), which is involved in the maintenance of cyto-architecture by interacting with actin-like filaments.


Subject(s)
Mutation/genetics , Proteins/genetics , Wiskott-Aldrich Syndrome/genetics , Adult , Base Sequence , Child , Child, Preschool , Codon, Nonsense/genetics , Frameshift Mutation/genetics , Genes, Recessive/genetics , Genotype , Humans , Infant , Male , Molecular Sequence Data , Phenotype , Polymerase Chain Reaction , Polymorphism, Single-Stranded Conformational , Proteins/chemistry , Sequence Homology, Nucleic Acid , Wiskott-Aldrich Syndrome Protein
15.
Am J Hum Genet ; 50(6): 1151-61, 1992 Jun.
Article in English | MEDLINE | ID: mdl-1598899

ABSTRACT

A locus for malignant hyperthermia susceptibility (MHS) has been localized on chromosome 19q12-13.2, while at the same time the gene encoding the skeletal muscle ryanodine receptor (RYR1) also has been mapped to this region and has been found to be tightly linked to MHS. RYR1 was consequently postulated as the candidate for the molecular defect causing MHS, and a point mutation in the gene has now been identified and is thought to be the cause of MH in at least some MHS patients. Here we report the results of a linkage study done with 19q12-13.2 markers, including the RYR1 cDNA, in two Bavarian families with MHS. In one of the families, three unambiguous recombination events between MHS and the RYR1 locus were found. In the second family only one informative meiosis was seen with RYR1. However, segregation analysis with markers for D19S75, D19S28, D19S47, CYP2A, BCL3, and APOC2 shows that the crossovers in the first family involve the entire haplotype defined by these markers flanking RYR1 and, furthermore, reveals multiple crossovers between these haplotypes and MHS in the second family. In these families, pairwise and multipoint lod scores below -2 exclude MHS from an interval spanning more than 26 cM and comprising the RYR1 and the previously described MHS locus. Our findings thus strongly suggest genetic heterogeneity of the MHS trait and prompt the search for another MHS locus.


Subject(s)
Chromosomes, Human, Pair 19 , Malignant Hyperthermia/genetics , Muscle Contraction , Muscles/physiopathology , Mutation , Receptors, Cholinergic/genetics , Adult , Child , Chromosome Banding , Chromosome Mapping , Female , Genetic Linkage , Genetic Markers , Genetic Predisposition to Disease , Humans , Male , Malignant Hyperthermia/physiopathology , Pedigree , Recombination, Genetic , Ryanodine/metabolism , Ryanodine Receptor Calcium Release Channel , Software
SELECTION OF CITATIONS
SEARCH DETAIL
...