Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
2.
Mutat Res ; 647(1-2): 86-93, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-18778718

ABSTRACT

Over the last several years there has been an explosion in our understanding of the organization of telomeric chromatin in mammals. As in other regions of the genome, chromatin composition at the telomere regulates structure, which defines function. Mammalian telomeres, similar to what has been demonstrated for telomeres of other eukaryotes, carry marks of heterochromatin and alteration in this underlying epigenetic code has effects on telomere replication and recombination. Experiments aimed at determining links between changes in telomeric chromatin and possible roles in aging and disease are beginning to emerge. The rapid refinement of our knowledge of the structure and alterations in telomeric chromatin over the last several years makes it likely that we are just seeing the tip of the iceberg.


Subject(s)
Aging/genetics , Genetic Diseases, Inborn/genetics , Neoplasms/genetics , Telomere/physiology , Animals , Chromatin/physiology , Mice
3.
Curr Mol Med ; 5(2): 187-96, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15974872

ABSTRACT

The use of chemotherapy provides an essential arm in the treatment of a number of cancers. The biological feature common to all cancerous cells that sensitizes them to chemotherapeutic agents is their elevated division rate. Rapidly dividing cells, such as tumor cells, are more sensitive to chemotherapeutic agents that act to initiate pathways leading to cell death, a process enhanced in cells with compromised DNA damage responses. The toxicity accompanying chemotherapy is due to side-effects induced in normal regenerative tissues which also have relatively high replication rates, such as hair follicles, the hematopoietic system, the gastrointestinal system, the germline and skin cells. While the side-effects of chemotherapy may be tolerated by the patient, the long term impact of the cytotoxic effects of chemotherapy on healthy tissues is only now becoming apparent. Chemotherapy-induced cytotoxicity in regenerative tissues requires multiple cell divisions in order to reconstitute the affected tissues. At least in part as a consequence of these extra divisions, telomeres in individuals treated with chemotherapy are shorter than age-matched control individuals who have never been exposed to these drugs. Given the essential role of telomeres in regulating cellular aging and chromosomal stability, it is possible that the prematurely shortened telomeres that arise following chemotherapy may impact the long-term replicative potential of these tissues. This review is focused on how telomeres may be modulated, directly or indirectly, by anticancer drugs and the potential long-term consequences of accelerated telomere shortening in healthy tissue as a result of current cancer treatment protocols.


Subject(s)
Neoplasms/drug therapy , Telomere/metabolism , Neoplasms/genetics
4.
Cytogenet Genome Res ; 105(2-4): 471-8, 2004.
Article in English | MEDLINE | ID: mdl-15237236

ABSTRACT

Telomeres are capping structures at the ends of chromosomes, composed of a repetitive DNA sequence and associated proteins. Both a minimal length of telomeric repeats and telomere-associated binding proteins are necessary for proper telomere function. Functional telomeres are essential for maintaining the integrity and stability of eukaryotic genomes. The capping structure enables cells to distinguish chromosome ends from double strand breaks (DSBs) in the genome. Uncapped chromosome ends are at great risk for degradation, recombination, or chromosome fusion by cellular DNA repair systems. Dysfunctional telomeres have been proposed to contribute to tumorigenesis and some aging phenotypes. The analysis of mice deficient in telomerase activity and other telomere-associated proteins has allowed the roles of dysfunctional telomeres in tumorigenesis and aging to be directly tested. Here we will focus on the analysis of different mouse models disrupted for proteins that are important for telomere functions and discuss known and proposed consequences of telomere dysfunction in tumorigenesis and aging.


Subject(s)
Aging/genetics , Cell Transformation, Neoplastic/genetics , Telomere , Animals , Antigens, Nuclear , Cyclin-Dependent Kinase Inhibitor p16/deficiency , DNA-Activated Protein Kinase , DNA-Binding Proteins/deficiency , Disease Models, Animal , Ku Autoantigen , Mice , Models, Animal , Protein Serine-Threonine Kinases/deficiency , RNA , Telomerase/deficiency , Telomerase/genetics , Tumor Suppressor Protein p14ARF/deficiency , Tumor Suppressor Protein p53/deficiency
5.
Bone Marrow Transplant ; 33(1): 71-8, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14704659

ABSTRACT

This study was conducted to determine if the accelerated telomere attrition that occurs as a consequence of allogeneic stem cell transplantation leads to measurable functional defects. Telomere lengths in mononuclear leukocytes obtained from 15 long-term allogeneic stem cell transplant recipients and their respective donors were determined by Southern hybridization and densitometric analysis. Functional assays evaluated the ability of these cells to proliferate in response to a mitogenic stimulus and to differentiate under appropriate cytokine stimulation. Lymphocyte proliferation in response to phytohemagglutinin was determined by measurement of (3)[H]thymidine uptake. The ability of circulating myeloid cells to differentiate was determined after incubation of peripheral blood mononuclear cells with IL-3 and GM-CSF. A total of 13 patients demonstrated telomeric loss, ranging from 0.1 to 3.7 kbp. Strikingly, lymphocytes from 14 of the 15 patients demonstrated a significant decrease in proliferation when compared to their respective donors (68%+/-22, P=0.001). All patients demonstrated at least a 50% decrease in the number of myeloid colony-forming units when compared to their respective donors (P<0.0001). A decreased ability of hematopoietic cells to proliferate and differentiate is phenotypically consistent with an aged immune system. This may correlate with diminished clinically relevant immune responses to infection or vaccination, as seen in the elderly.


Subject(s)
Bone Marrow Transplantation/standards , Immune System/physiopathology , Peripheral Blood Stem Cell Transplantation/standards , Telomere/immunology , Adult , Aged , Cell Differentiation/drug effects , Cellular Senescence , Cytokines/pharmacology , Female , Hematopoiesis , Humans , Immune System/cytology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/physiology , Lymphocyte Activation/drug effects , Male , Middle Aged , Myeloid Cells/cytology , Myeloid Progenitor Cells , Myelopoiesis , Phytohemagglutinins/pharmacology , Survivors , Telomere/ultrastructure , Transplantation, Homologous
6.
Hum Mol Genet ; 10(18): 1953-61, 2001 Sep 01.
Article in English | MEDLINE | ID: mdl-11555632

ABSTRACT

Telomere length maintenance is essential for cellular immortalization, and thus tumorigenesis. Most human tumors and immortal cell lines maintain their telomeric DNA via the activity of a specialized reverse transcriptase, telomerase. Stabilization of telomeric repeat tracts may also be achieved through a telomerase-independent mechanism, referred to as alternative lengthening of telomeres (ALT). ALT cells are telomerase negative and are characterized by extremely long and heterogeneously sized telomeres and novel multiprotein structures called ALT-associated PML nuclear bodies which are unique to ALT cells. To determine if reconstitution of telomerase activity suppressed ALT and restored wild-type telomere lengths, we introduced the catalytic subunit of telomerase into two ALT cell lines. Initially, two clonal lines exhibited enrichment of shorter telomeres while maintaining a population of ultra-long telomeres similar to that observed in the parental line, suggesting that telomerase is stabilizing the shorter telomeres in the population. Telomere length in the third clonal line was not detectably different from that in the parental cell line. One clonal line with a phenotype of shorter telomeres maintained this pattern over time in culture while the second gradually reverted to the parental ALT telomere length pattern, concurrent with reduction of telomerase activity. All clones continued to maintain ALT-associated PML nuclear bodies regardless of whether telomerase was present. The data suggest that introduction of telomerase activity alone is not sufficient to completely repress ALT, that telomerase acts preferentially on the shortest telomeres in the culture and that the ALT and telomerase pathways may be present concurrently in mammalian cells.


Subject(s)
Nuclear Proteins , Recombination, Genetic , Telomerase/metabolism , Telomere/genetics , Blotting, Southern , Catalytic Domain , Cell Line , Cell Nucleus/metabolism , Clone Cells , DNA/genetics , DNA-Binding Proteins/analysis , Female , Fluorescent Antibody Technique, Indirect , G2 Phase , Humans , Mitosis , Neoplasm Proteins/analysis , Promyelocytic Leukemia Protein , Telomerase/genetics , Telomere/metabolism , Transcription Factors/analysis , Transfection , Tumor Suppressor Proteins
8.
J Cell Sci ; 113 Pt 24: 4577-85, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11082050

ABSTRACT

Telomere maintenance is essential for the unlimited proliferative potential of human cells, and hence immortalization. However, a number of tumors, tumor-derived cell lines and in vitro immortalized cell lines have been described that do not express detectable telomerase activity. These lines utilize a mechanism, termed Alternative Lengthening of Telomeres (ALT), to provide telomere maintenance. A subset of the cells in each ALT cell line contain a novel form of the promyelocytic leukemia nuclear body (PML NB) in which telomeric DNA and the telomere binding proteins TRF1 and TRF2 co-localize with the PML protein, termed ALT-associated PML bodies (AA-PBs). In contrast, in non-ALT, telomerase-positive cell lines these telomeric proteins and the PML NB occupy distinct and separate subnuclear domains. PML NBs have been implicated in terminal differentiation, growth suppression and apoptosis. The role, if any, of AA-PBs in telomere maintenance or culture viability in telomerase negative cell lines is unclear, but it has been suggested that cells containing these structures are no longer viable and are marked for eventual death. We utilized a series of human ovarian surface epithelium (HOSE) cell lines that use ALT for telomere maintenance to determine if AA-PBs are indeed markers of cells in these cultures that are no longer cycling. We show that AA-PB positive cells incorporate BrdU and thus are able to carry out DNA replication. In addition, AA-PBs are present in mitotic cells and the frequency of cells containing these structures is increased when cultures are enriched for cells in the G(2)/M phase of the cell cycle suggesting that the formation of AA-PBs is coordinately regulated with the cell cycle. Finally, we demonstrate that the majority of the AA-PB positive cells in the culture are not destined for immediate apoptosis. Taken together the data argue against AA-PBs marking cells destined for death and, instead, raise the possibility that these structures may be actively involved in telomere maintenance via the ALT pathway.


Subject(s)
Neoplasm Proteins/metabolism , Nuclear Proteins , Telomere/physiology , Transcription Factors/metabolism , Cell Cycle , Cell Line , Cell Survival , DNA-Binding Proteins/metabolism , Epithelial Cells , Female , G2 Phase , HeLa Cells , Humans , Mitosis/physiology , Ovary/cytology , Promyelocytic Leukemia Protein , Telomeric Repeat Binding Protein 1 , Tumor Suppressor Proteins
10.
Science ; 283(5406): 1321-5, 1999 Feb 26.
Article in English | MEDLINE | ID: mdl-10037601

ABSTRACT

Although broken chromosomes can induce apoptosis, natural chromosome ends (telomeres) do not trigger this response. It is shown that this suppression of apoptosis involves the telomeric-repeat binding factor 2 (TRF2). Inhibition of TRF2 resulted in apoptosis in a subset of mammalian cell types. The response was mediated by p53 and the ATM (ataxia telangiectasia mutated) kinase, consistent with activation of a DNA damage checkpoint. Apoptosis was not due to rupture of dicentric chromosomes formed by end-to-end fusion, indicating that telomeres lacking TRF2 directly signal apoptosis, possibly because they resemble damaged DNA. Thus, in some cells, telomere shortening may signal cell death rather than senescence.


Subject(s)
Apoptosis , DNA-Binding Proteins/physiology , Protein Serine-Threonine Kinases , Telomere/physiology , Tumor Suppressor Protein p53/metabolism , Adenoviridae/genetics , Adenoviridae/physiology , Animals , Ataxia Telangiectasia/pathology , Ataxia Telangiectasia Mutated Proteins , B-Lymphocytes/cytology , Cell Cycle Proteins , Cell Line , Cells, Cultured , Cloning, Molecular , DNA Damage , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Genetic Vectors , Humans , In Situ Nick-End Labeling , Mice , Mitosis , Phosphorylation , Proteins/metabolism , T-Lymphocytes/cytology , Telomeric Repeat Binding Protein 2 , Tumor Cells, Cultured , Tumor Suppressor Proteins
11.
Nat Genet ; 17(2): 231-5, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9326950

ABSTRACT

Human telomeres are composed of long arrays of TTAGGG repeats that form a nucleoprotein complex required for the protection and replication of chromosome ends. One component of human telomeres is the TTAGGG repeat binding factor 1 (TRF1), a ubiquitously expressed protein, related to the protooncogene Myb, that is present at telomeres throughout the cell cycle. Recent evidence has implicated TRF1 in the control of telomere length. TRF1 is proposed to be an inhibitor of telomerase, acting in cis to limit the elongation of individual chromosome ends. Here we report the cloning of TRF2, a distant homologue of TRF1 that carries a very similar Myb-related DNA-binding motif. Like TRF1, TRF2 was ubiquitously expressed, bound specifically to duplex TTAGGG repeats in vitro and localized to all human telomeres in metaphase chromosomes. TRF2 was shown to have an architecture similar to that of TRF1 in that it carries a C-terminal Myb motif and a large TRF1-related dimerization domain near its N terminus. However, the dimerization domains of TRF1 and TRF2 did not interact, suggesting that these proteins exist predominantly as homodimers. While having similar telomere binding activity and domain organization, TRF2 differed from TRF1 in that its N terminus was basic rather than acidic, and TRF2 was much more conserved than TRF1. The results indicate that the TTAGGG repeat arrays at the ends of human and mouse chromosomes bind to two related proteins. Because TRF1 and TRF2 showed significant differences, we suggest that these factors have distinct functions at telomeres.


Subject(s)
DNA-Binding Proteins/metabolism , Telomere/metabolism , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Cloning, Molecular , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Dimerization , HeLa Cells , Humans , In Vitro Techniques , Mice , Microsatellite Repeats , Molecular Sequence Data , Oligopeptides , Peptides/metabolism , Protein Binding , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-myb , Telomere/genetics , Telomeric Repeat Binding Protein 1 , Telomeric Repeat Binding Protein 2 , Trans-Activators/genetics , Trans-Activators/metabolism , Transfection
12.
Hum Mol Genet ; 6(1): 69-76, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9002672

ABSTRACT

Mammalian chromosome ends contain long arrays of TTAGGG repeats that are complexed to a telomere specific protein, the TTAGGG repeat binding factor, TRF1. Here we describe the characterization of genes encoding the human and mouse TRF1 proteins, hTRF1 and mTRF1. The mTRF1 cDNA was isolated based on sequence similarity to the hTRF1 cDNA and the mTRF1 mRNA was shown to be ubiquitously expressed as a single 1.9 kb polyadenylated transcript in mouse somatic tissues. High levels of a 2.1 kb transcript were found in testes. In vitro translation of the mTRF1 cDNA resulted in a 56 kDa protein that binds to TTAGGG repeat arrays. mTRF1 displayed the same sequence specificity as hTRF1, preferring arrays of TTAGGG repeats as a binding substrate over TTAGGC and TTGGGG repeats. Expression of an epitope-tagged version of mTRF1 showed that the protein is located at the ends of murine metaphase chromosomes. In agreement, conceptual translation indicated that mTRF1 and hTRF1 are similarly-sized proteins with nearly identical C-terminal Myb-related DNA binding motifs. In addition, comparison of the predicted mTRF1 and hTRF1 amino acid sequences showed that the acidic nature of the N-terminus of TRF1 is conserved and revealed a highly conserved novel domain of approximately 200 amino acids in the middle of the proteins. However, other regions of the proteins are poorly conserved (<35% identity) and the overall level of identity of the mTRF1 and hTRF1 amino acid sequences is only 67%. The TRF1 genes are not syntenic; the hTRF1 gene localized to human chromosome 8 band q13 while the mTRF1 gene localized to mouse chromosome 17 band E3. The data indicate that the genes for mammalian telomeric proteins evolve rapidly.


Subject(s)
Chromosome Mapping , Conserved Sequence , DNA-Binding Proteins/genetics , Gene Expression , Amino Acid Sequence , Animals , Base Sequence , Chromosomes , DNA, Complementary , DNA-Binding Proteins/metabolism , Genes , Germ Cells , Humans , Metaphase , Mice , Molecular Sequence Data , Open Reading Frames , Sequence Homology, Amino Acid , Telomere , Telomeric Repeat Binding Protein 1 , Tissue Distribution
13.
Mol Cell Biol ; 16(7): 3765-72, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8668193

ABSTRACT

Activation of telomerase in human cancers is thought to be necessary to overcome the progressive loss of telomeric DNA that accompanies proliferation of normal somatic cells. According to this model, telomerase provides a growth advantage to cells in which extensive terminal sequence loss threatens viability. To test these ideas, we have examined telomere dynamics and telomerase activation during mammary tumorigenesis in mice carrying a mouse mammary tumor virus long terminal repeat-driven Wnt-1 transgene. We also analyzed Wnt-1-induced mammary tumors in mice lacking p53 function. Normal mammary glands, hyperplastic mammary glands, and mammary carcinomas all had the long telomeres (20 to 50 kb) typical of Mus musculus and did not show telomere shortening during tumor development. Nevertheless, telomerase activity and the RNA component of the enzyme were consistently upregulated in Wnt-1-induced mammary tumors compared with normal and hyperplastic tissues. The upregulation of telomerase activity and RNA also occurred during tumorigenesis in p53-deficient mice. The expression of telomerase RNA correlated strongly with histone H4 mRNA in all normal tissues and tumors, indicating that the RNA component of telomerase is regulated with cell proliferation. Telomerase activity in the tumors was elevated to a greater extent than telomerase RNA, implying that the enzymatic activity of telomerase is regulated at additional levels. Our data suggest that the mechanism of telomerase activation in mouse mammary tumors is not linked to global loss of telomere function but involves multiple regulatory events including upregulation of telomerase RNA in proliferating cells.


Subject(s)
Mammary Neoplasms, Experimental/enzymology , Proto-Oncogene Proteins/genetics , Telomerase/metabolism , Zebrafish Proteins , Animals , Base Sequence , Enzyme Activation , Female , Gene Expression Regulation, Neoplastic , Histones/biosynthesis , Humans , Hyperplasia , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/pathology , Mammary Tumor Virus, Mouse , Mice , Mice, Transgenic , Mitogens/genetics , Molecular Sequence Data , Oligodeoxyribonucleotides , Polymerase Chain Reaction , Proto-Oncogene Proteins/biosynthesis , RNA Polymerase III/metabolism , RNA, Messenger/biosynthesis , Repetitive Sequences, Nucleic Acid , Telomerase/biosynthesis , Transcription, Genetic , Wnt Proteins , Wnt1 Protein
14.
Science ; 270(5242): 1663-7, 1995 Dec 08.
Article in English | MEDLINE | ID: mdl-7502076

ABSTRACT

Telomeres are multifunctional elements that shield chromosome ends from degradation and end-to-end fusions, prevent activation of DNA damage checkpoints, and modulate the maintenance of telomeric DNA by telomerase. A major protein component of human telomeres has been identified and cloned. This factor, TRF, contains one Myb-type DNA-binding repeat and an amino-terminal acidic domain. Immunofluorescent labeling shows that TRF specifically colocalizes with telomeric DNA in human interphase cells and is located at chromosome ends during metaphase. The presence of TRF along the telomeric TTAGGG repeat array demonstrates that human telomeres form a specialized nucleoprotein complex.


Subject(s)
DNA-Binding Proteins/chemistry , Telomere/chemistry , Amino Acid Sequence , Base Sequence , Cell Nucleus/chemistry , Cloning, Molecular , DNA-Binding Proteins/analysis , DNA-Binding Proteins/genetics , DNA-Binding Proteins/isolation & purification , HeLa Cells , Humans , Interphase , Metaphase , Molecular Sequence Data , Molecular Weight , Repetitive Sequences, Nucleic Acid , Sequence Alignment , Transfection
15.
Proc Natl Acad Sci U S A ; 92(20): 9082-6, 1995 Sep 26.
Article in English | MEDLINE | ID: mdl-7568077

ABSTRACT

Bone marrow and peripheral blood leukocytes from 19 leukemia patients were found to contain telomerase activity detectable by a PCR-based assay. Telomerase was also detectable in nonmalignant bone marrow and peripheral blood leukocytes from normal donors, including fractions enriched for granulocytes, T lymphocytes, and monocytes/B cells. Semiquantitative comparison revealed considerable overlap between telomerase activities in samples from normal subjects and leukemia patients, confounding evaluation of the role of telomerase in this disease. These data indicate that human telomerase is not restricted to immortal cells and suggest that the somatic expression of this enzyme may be more widespread than was previously inferred from the decline of human telomeres.


Subject(s)
Granulocytes/enzymology , Hematopoietic Stem Cells/enzymology , Leukemia/enzymology , Lymphocytes/enzymology , Telomerase/metabolism , Acute Disease , B-Lymphocytes/enzymology , Base Sequence , Bone Marrow/enzymology , Bone Marrow/pathology , DNA Primers , Humans , Kinetics , Leukemia/blood , Leukemia/pathology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Molecular Sequence Data , Polymerase Chain Reaction/methods , Reference Values , Remission Induction , T-Lymphocytes/enzymology , Telomerase/analysis , Telomerase/blood
16.
Chromosoma ; 103(10): 685-92, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7664615

ABSTRACT

A combination of cytogenetic and molecular biology techniques were used to study the molecular composition and organisation of the pericentromeric regions of house mouse metacentric chromosomes, the products of Robertsonian (Rb) translocations between telocentrics. Regardless of whether mitotic or meiotic preparations were used, in situ hybridisation failed to reveal pericentromeric telomeric sequences on any of the Rb chromosomes, while all metacentrics retained detectable, although reduced (average 50 kb), amounts of minor satellite DNA in the vicinity of their centromeres. These results were supported by slot blot hybridisation which indicated that mice with 2n=22 Rb chromosomes have 65% of telomeric sequences (which are allocated to the distal telomeres of both Rb and telocentric chromosomes and to the proximal telomeres of telocentrics) and 15% the amount of minor satellite, compared with mice with 2n=40 all-telocentric chromosomes. Pulsed field gel electrophoresis and Southern analysis of DNA from Rb mice showed that the size of the telomeric arrays is similar to that of mice with all-telocentric chromosomes and that the minor satellite sequences were hybridising to larger fragments incorporating major satellite DNA. Since the telomeric sequences are closer to the physical end of the chromosome than the minor satellite sequences, the absence of telomeric sequences and the reduced amount of minor satellite sequences at the pericentromeric region of the Rb metacentrics suggest that the breakpoints for the Rb translocation occur very close to the minor satellite-major satellite border. Moreover, it is likely that the minor satellite is required for centromeric function, 50-67 kb being enough DNA to organise one centromere with a functionally active kinetochore.


Subject(s)
Centromere , DNA, Satellite , Telomere , Translocation, Genetic , Animals , DNA Probes , Electrophoresis, Gel, Pulsed-Field , Female , In Situ Hybridization , Male , Mice , Mice, Inbred C3H
17.
Exp Cell Res ; 212(2): 308-13, 1994 Jun.
Article in English | MEDLINE | ID: mdl-8187823

ABSTRACT

Chromosomes occupy specific nonrandom domains in the interphase nucleus of eukaryotic cells. We have used a Chinese hamster-human somatic cell hybrid line containing a single human X chromosome to study the interphase distribution of the Xp telomere using fluorescent in situ hybridization and optical sectioning. A derivative cell line in which the X chromosome has been broken at Xq22-24 and healed by the addition of cloned human telomeric sequences was also studied to determine if introduction of these sequences at a previously interstitial site changed its location in interphase. The endogenous Xp telomere occupies a specific, nonrandom, internal domain. Introduction of a telomere at a previously interstitial site did not alter the interphase nuclear location of that site. The results suggest that nonrandom interphase location of telomeres may not be determined solely by the DNA sequence of the telomere.


Subject(s)
Cell Nucleus/ultrastructure , Interphase , Telomere , X Chromosome/ultrastructure , Humans , Hybrid Cells
18.
Hum Mol Genet ; 3(4): 539-46, 1994 Apr.
Article in English | MEDLINE | ID: mdl-8069295

ABSTRACT

Pericentric heterochromatin and telomeres have been shown to be capable of repressing the expression of genes located in close proximity. The effect of adjacent structural sequences on gene expression will be important in the design of mammalian artificial chromosomes. In the process of using telomere-containing constructs to generate a deletion panel of the long arm of the human X chromosome, several cell lines were produced which appeared by in situ hybridization to be broken in Xq at or near the centromere. After analysis of end clones rescued from these cell lines, only two produced data consistent with breaks in the alpha satellite array without accompanying rearrangements. The mitotic stability of an X chromosome, with at least 750 kb of the alpha satellite array deleted, was compared to controls where the alpha satellite array remained intact. No significant change in the stability of the chromosome was observed, suggesting that the truncated chromosome has a fully functional mitotic centromere. There was no detectable change in the expression of the hygromycin resistance gene, which is located between a functional centromere and telomere, in this cell line. This study indicates that structural elements flanking a mammalian selectable marker do not result in silencing.


Subject(s)
Cinnamates , DNA, Satellite/genetics , Gene Expression Regulation , Heterochromatin , Telomere , X Chromosome , Animals , CHO Cells , Cell Line , Cricetinae , Drug Resistance , Electrophoresis, Gel, Pulsed-Field , Humans , Hybrid Cells , Hygromycin B/analogs & derivatives , Hygromycin B/pharmacology , In Situ Hybridization, Fluorescence , Molecular Sequence Data , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Sequence Deletion
20.
Chromosoma ; 101(7): 442-7, 1992 Apr.
Article in English | MEDLINE | ID: mdl-1618025

ABSTRACT

A mouse subtelomeric sequence, ST1, was generated from genomic DNA of the mouse HR9 (129/Sv origin) cell line by the polymerase chain reaction (PCR) using a single telomeric primer. ST1 was cloned and characterized: it is composed of 670 bp of novel DNA sequence flanked on each end by inverted telomeric hexanucleotide repeats (TTAGGG)n. PCR amplification from BALB/c mouse DNA using this single primer gave the same major product. Southern analysis and PCR using internal ST1 primers confirmed that the ST1 sequence is present in mouse genomic DNA. In situ hybridization to metaphase chromosomes of SJL origin mapped ST1 to many, if not every, mouse telomere. PCR experiments using different combinations of the telomeric, minor satellite, and ST1 primers indicated that some ST1 copies are adjacent to minor satellite sequences, that telomeric and ST1 sequences are not generally interspersed with minor satellite sequences, and that ST1 and the minor satellite have a consistent and specific orientation relative to each other and to the telomere.


Subject(s)
DNA/isolation & purification , Telomere/chemistry , Animals , Base Sequence , Blotting, Southern , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...