Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 9 de 9
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Cancer Res ; 61(20): 7594-602, 2001 Oct 15.
Article de Anglais | MEDLINE | ID: mdl-11606399

RÉSUMÉ

Telomerase is required for the complete replication of chromosomal ends. In tumors, the human telomerase reverse transcriptase subunit (hTERT) is up-regulated, thereby removing a critical barrier for unlimited cell proliferation. To understand more about hTERT regulation, we measured hTERT RNA levels by quantitative reverse transcription (RT)-PCR. Telomerase-positive cell lines were found to contain between 0.2 and 6 molecules of spliced hTERT RNA per cell, whereas in telomerase-negative cells, the number of molecules was below the sensitivity of the assay (<0.004 molecules/cell). Intron-containing, immature hTERT RNA was observed only in nuclei of telomerase-positive cells, which suggests that hTERT RNA levels are transcriptionally regulated. Microcell transfer of a normal chromosome 3 into the human breast carcinoma cell line (21NT) abolishes telomerase activity and induces senescence. Endogenous hTERT transcripts were undetectable in the nuclei of 21NT-chromosome 3 hybrids, even in cells permanently expressing a transfected hTERT cDNA. However, chromosome 3 transfer did not affect the expression of green fluorescent protein reporter constructs driven by up to 7.4 kb of noncoding DNA flanking the 5' end of the hTERT gene. Because direct up-regulation of hTERT through c-Myc overexpression had previously been reported, we investigated whether chromosome 3 transfer affected c-Myc activity. An at least 30-fold reduction of immature intron-containing hTERT RNA was observed after the introduction of a normal chromosome 3, but expression levels of c-Myc, Mad1, and other c-Myc target genes were unchanged. Our results suggest that telomerase is regulated primarily at the level of hTERT transcription by complex mechanisms involving regulatory elements distant from the 5' flanking region, and that the putative hTERT repressor on chromosome 3 does not regulate the expression of hTERT through c-Myc or one of its coregulators.


Sujet(s)
Chromosomes humains de la paire 3/physiologie , Régulation de l'expression des gènes au cours du développement , Protéines proto-oncogènes c-myc/physiologie , ARN messager/antagonistes et inhibiteurs , Telomerase/antagonistes et inhibiteurs , Telomerase/métabolisme , Tumeurs du sein/enzymologie , Tumeurs du sein/génétique , Cycle cellulaire/génétique , Différenciation cellulaire/génétique , Noyau de la cellule/génétique , Noyau de la cellule/métabolisme , Protéines de liaison à l'ADN , Régulation négative , Fibroblastes/enzymologie , Régulation de l'expression des gènes tumoraux , Humains , Protéines proto-oncogènes c-myc/génétique , Épissage des ARN , ARN messager/biosynthèse , ARN messager/génétique , ARN tumoral/génétique , ARN tumoral/métabolisme , Telomerase/biosynthèse , Telomerase/génétique , Cellules cancéreuses en culture
2.
J Natl Cancer Inst ; 93(11): 865-72, 2001 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-11390536

RÉSUMÉ

BACKGROUND: High-risk human papillomavirus (HPV) types play a major role in the development of cervical cancer in vivo and can induce immortalization of primary human keratinocytes in vitro. Activation of the telomere-lengthening enzyme telomerase constitutes a key event in both processes. Because losses of alleles from chromosome 6 and increased telomerase activity have been observed in high-grade premalignant cervical lesions, we analyzed whether human chromosome 6 harbors a putative telomerase repressor locus that may be involved in HPV-mediated immortalization. METHODS: Microcell-mediated chromosome transfer was used to introduce chromosomes 6 and 11 to the in vitro generated HPV type 16 (HPV16)-immortalized keratinocyte cell line FK16A and to the in vivo derived HPV16-containing cervical cancer cell line SIHA: Hybrid clones were analyzed for growth characteristics, telomerase activity, human telomerase reverse transcriptase (hTERT) and HPV16 E6 expression, and telomere length. FK16A hybrid clones were also transduced with an hTERT-containing retrovirus to examine the effect of ectopic hTERT expression on growth. Statistical tests were two-sided. RESULTS: Introduction of human chromosome 6 but not of chromosome 11 to both cell lines yielded hybrid cells that demonstrated crisis-like features (i.e., enlarged and flattened morphology, vacuolation, and multinucleation) and underwent growth arrest after a marked lag period. In the chromosome 6 hybrid clones analyzed, telomerase activity and hTERT messenger RNA (mRNA) expression were statistically significantly reduced compared with those in the chromosome 11 hybrid clones (for telomerase activity, P =.004 for the FK16A hybrids and P =.039 for the SiHa hybrids; for hTERT mRNA expression, P =.003 for the FK16A hybrids). The observed growth arrest was associated with telomeric shortening. Ectopic expression of hTERT in FK16A cells could prevent the telomeric shortening-based growth arrest induced by chromosome 6. CONCLUSIONS: Chromosome 6 may harbor a repressor of hTERT transcription, the loss of which may be involved in HPV-mediated immortalization.


Sujet(s)
Chromosomes humains de la paire 6 , Papillomaviridae/génétique , ARN , Telomerase/métabolisme , Tumeurs du col de l'utérus/génétique , Division cellulaire , Lignée de cellules transformées , Chromosomes humains de la paire 11 , Protéines de liaison à l'ADN , Femelle , Gènes rapporteurs , Humains , Cellules hybrides , Kératinocytes , Répétitions microsatellites , Réaction de polymérisation en chaîne , RT-PCR , Telomerase/antagonistes et inhibiteurs , Télomère/génétique , Télomère/ultrastructure , Transfection , Cellules cancéreuses en culture , beta-Galactosidase/génétique
3.
J Natl Cancer Inst ; 91(1): 37-45, 1999 Jan 06.
Article de Anglais | MEDLINE | ID: mdl-9890168

RÉSUMÉ

BACKGROUND: Activation of the enzyme telomerase, which has been associated with cellular immortality, may constitute a key step in the development of human cancer. Telomerase is repressed in most normal human somatic cells. This study was conducted, using a genetic complementation approach, with the aim of identifying and mapping the genes responsible for repressing telomerase and, simultaneously, to establish the effect of experimentally induced telomerase repression on human tumor cell growth. METHODS: Individual human chromosomes isolated from normal diploid cells and tagged with bacterial antibiotic resistance genes (for later selection) were introduced into cells of the human breast carcinoma cell line 21NT by means of microcell transfer. Selected hybrid clones were screened for telomerase activity by use of the polymerase chain reaction-based telomere repeat amplification protocol (TRAP) assay, and the proliferative fate of the hybrid clones was determined. Regions of the introduced chromosomes associated with telomerase repression were mapped using segregant hybrids and a deletion analysis that employed microsatellite DNA markers. RESULTS: Strong repression of telomerase was observed following transfer of human chromosome 3 into 21NT cells but not after transfer of chromosomes 8, 12, or 20. The vast majority of hybrid clones with repressed telomerase entered permanent growth arrest after 10-18 population doublings. Deletion analysis of nonrepressed segregant monochromosome 3 hybrids indicated two regions on the short arm of chromosome 3 (3p21.3-p22 and 3p12-21.1) where telomerase regulator genes may be located. CONCLUSIONS: Telomerase in human breast cancer cells is efficiently repressed by a gene or genes on normal human chromosome 3p, and this repression is associated with permanent growth arrest of the tumor cells.


Sujet(s)
Tumeurs du sein/anatomopathologie , Carcinome intracanalaire non infiltrant/anatomopathologie , Chromosomes humains de la paire 3/génétique , Régulation de l'expression des gènes codant pour des enzymes , Techniques de transfert de gènes , Inhibiteurs de croissance/génétique , Protéines tumorales/biosynthèse , Protéines de répression/génétique , Telomerase/biosynthèse , Adulte , Carcinome intracanalaire non infiltrant/enzymologie , Division cellulaire/effets des médicaments et des substances chimiques , Fusion cellulaire , Chromosomes/métabolisme , Chromosomes/ultrastructure , Femelle , Inhibiteurs de croissance/physiologie , Humains , Cellules hybrides , Hybridation fluorescente in situ , Protéines tumorales/génétique , Protéines tumorales/physiologie , Réaction de polymérisation en chaîne , Protéines de répression/physiologie , Telomerase/génétique , Telomerase/physiologie , Télomère/métabolisme , Cellules cancéreuses en culture
4.
Oncogene ; 17(26): 3417-26, 1998 Dec 31.
Article de Anglais | MEDLINE | ID: mdl-10030665

RÉSUMÉ

Reactivation of telomerase and stabilization of telomeres occur simultaneously during human cell immortalization in vitro and the vast majority of human cancers possess high levels of telomerase activity. Telomerase repression in human somatic cells may therefore have evolved as a powerful resistance mechanism against immortalization, clonal evolution and malignant progression. The comparative ease with which rodent cells immortalize in vitro suggests that they have less stringent controls over replicative senescence than human cells. Here, we report that Syrian hamster dermal fibroblasts possess substantial levels of telomerase activity throughout their culture life-span, even after growth arrest in senescence. In our studies, telomerase was also detected in uncultured newborn hamster skin, in several adult tissues, and in cultured fibroblasts induced to enter the post-mitotic state irreversibly by serum withdrawal. Transfection of near-senescent dermal fibroblasts with a selectable plasmid vector expressing the SV40 T-antigen gene resulted in high-frequency single-step immortalization without the crisis typically observed during the immortalization of human cells. Collectively, these data provide an explanation for the increased susceptibility of rodent cells to immortalization (and malignant transformation) compared with their human equivalents, and provide evidence for a novel, growth factor-sensitive, mammalian senescence mechanism unrelated to telomere maintenance.


Sujet(s)
Vieillissement de la cellule/physiologie , Telomerase/métabolisme , Télomère/physiologie , Animaux , Antigènes transformants de polyomavirus/génétique , Division cellulaire , Lignée de cellules transformées , Cellules cultivées , Cricetinae , Milieux de culture sans sérum , Facteurs temps
5.
Carcinogenesis ; 17(8): 1567-75, 1996 Aug.
Article de Anglais | MEDLINE | ID: mdl-8761411

RÉSUMÉ

Microcell transfer of intact normal human chromosomes into immortal mouse and hamster fibroblast cell lines has revealed growth suppressive activity associated with a small sub-set of the human complement. Here, we describe the results of a detailed study aimed at identifying the gene or genes responsible for the rapid growth-arrest response obtained with human chromosome-9. Initially, STS-PCR deletion mapping of segregants arising in monochromosome transfer experiments was used successfully to localize the active sub-chromosomal region to 9p21. Subsequent fine-structure deletion mapping of previously uniformative hybrid segregants, employing additional markers between D9S162 and D9S171, provided strong evidence that the cyclin-dependent kinase (cdk) inhibitor gene CDKN2A (p16INK4A) was solely responsible for the chromosome-9 effect; 9p21 microdeletions in a significant proportion of segregant clones were restricted to a single CDKN2A exon. Transfection experiments with CDKN2A and CDKN2B cDNA expression vectors, using mouse A9 cells and three human malignant melanoma cell lines as recipients, provided further evidence in support of this hypothesis. Collectively, our results indicate that expression of human CDKN2A (controlled either by its natural regulatory elements, or by a cytomegalovirus promoter) is incompatible with in vitro proliferation in immortalized rodent cells and in human melanoma cell lines. The rapidity of the growth inhibitory effects of CDKN2A was inconsistent with a mode of action involving induction of replicative cell senescence via telomerase repression, but was consistent with a mechanism based on cell cycle arrest through cdk inhibition. The study described here has generated a panel of microdeleted monochromosome-9 donor hybrids which may prove valuable in functional investigations aimed at identifying other important tumour suppressor genes located on human chromosome-9.


Sujet(s)
Protéines de transport/génétique , Chromosomes humains de la paire 9 , Gènes suppresseurs de tumeur , Animaux , Séquence nucléotidique , Division cellulaire/génétique , Lignée de cellules transformées , Inhibiteur p16 de kinase cycline-dépendante , Amorces ADN , Marqueurs génétiques , Humains , Cellules hybrides , Mélanome/génétique , Données de séquences moléculaires , Transfection , Cellules cancéreuses en culture
6.
Mol Carcinog ; 12(4): 213-24, 1995 Apr.
Article de Anglais | MEDLINE | ID: mdl-7727043

RÉSUMÉ

Recent demonstrations of loss of heterozygosity in a wide variety of human cancers suggest that large multilocus genetic deletions (presumably including tumor suppressor genes) constitute a major class of genetic alteration in human carcinogenesis. Here we show that a bifunctional fusion gene (Hytk), suitable for both positive and negative selection, is an effective marker for studying genetic loss in mammalian cells with minimal interference from point-mutational changes. Studies with a transgenic V79 cell line in which a single functional copy of Hytk was stably inserted into the genome in a retroviral vector showed that loss of the marker (and presumably flanking cellular genetic material) could be induced efficiently by ionizing radiation (gamma-rays and fast neutrons) but only weakly by the powerful point-mutagen benzo[a]pyrene diol epoxide. In a first application of the system, we provide evidence that radiation-induced loss can occur through an indirect mechanism after a high-frequency event. Collectively, our results suggest that the Hytk marker should be a valuable tool for studying genome position effects on the tolerance of genetic loss in cultured human cells that represent different stages in clonal evolution and tumor progression.


Sujet(s)
Délétion de segment de chromosome , Clonage moléculaire , Délétion de gène , Marqueurs génétiques , Cellules 3T3 , 7,8,8a,9a-Tétrahydro-benzo[10,11]chryséno[3,4-b]oxirène-7,8-diol/toxicité , Animaux , Lignée cellulaire , Survie cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des radiations , Relation dose-effet des rayonnements , Gènes suppresseurs de tumeur , Variation génétique , Humains , Souris , Neutrons , Plasmides , Réaction de polymérisation en chaîne , Provirus , Retroviridae , Transfection
7.
Carcinogenesis ; 16(2): 193-204, 1995 Feb.
Article de Anglais | MEDLINE | ID: mdl-7859348

RÉSUMÉ

Immortalization is a prerequisite for the clonal evolution and malignant transformation of normal mammalian cells in culture. In order to gain a mechanistic insight into the genetics of carcinogen-induced cellular immortality, a cell culture assay has been developed based on the use of freshly explanted Syrian hamster dermal (SHD) fibroblasts. The relative efficacies of a variety of chemical and physical carcinogens at immortalizing SHD cells (against a zero background of spontaneous immortalization) were compared. Ionizing radiation and nickel chloride appeared to be more effective as immortalizing agents than powerful point mutagens, suggesting (but not proving) that clastogenic damage may be more significant in the immortalization process than point mutation. Frequencies of induced immortality (10(-6)-10(-7)/treated cell) were arguably consistent with a direct mutational mechanism involving a single genetic target. However, detailed cytogenetic characterization of a panel of newly immortalized cell lines revealed no non-random chromosomal alterations in the cells at the level of G-banding. Furthermore, additional experiments with the SHD system have provided confirmatory evidence that immortalization can occur as an indirect consequence of carcinogen exposure following an induced high frequency change in the treated population, rather than through direct targeted mutagenesis. Previous somatic cell genetic studies have suggested the possibility that a target gene for immortalization exists on the human and Chinese hamster X chromosomes. Here we provide strong evidence that the normal SHD X chromosome displays powerful senescence-inducing properties when introduced, by microcell transfer, into newly immortalized SHD recipients. These results suggest that induction of the immortal phenotype in SHD cells by carcinogens results primarily from functional inactivation of a senescence gene which may be X-linked. One possible mechanism for senescence gene inactivation consistent with our observations is through a sub-microscopic interstitial genetic deletion. However, the considerable efficacy of nickel (a human carcinogen) as an immortalizing agent at nonmutagenic doses raises the alternative possibility that immortalization may occur through an epigenetic mechanism.


Sujet(s)
Cancérogènes/toxicité , Transformation cellulaire néoplasique , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/effets des radiations , Peau/effets des médicaments et des substances chimiques , Peau/effets des radiations , Animaux , Transformation cellulaire néoplasique/génétique , Chromosomes/effets des médicaments et des substances chimiques , Chromosomes/effets des radiations , Clones cellulaires , Cricetinae , Cricetulus , Relation dose-effet des médicaments , Relation dose-effet des rayonnements , Fibroblastes/cytologie , Techniques de transfert de gènes , Gènes/effets des médicaments et des substances chimiques , Gènes/effets des radiations , Humains , Caryotypage , Mesocricetus , Souris , Phénotype , Rats , Peau/cytologie , Transformation génétique
8.
Cytogenet Cell Genet ; 71(1): 68-76, 1995.
Article de Anglais | MEDLINE | ID: mdl-7606932

RÉSUMÉ

Human:rodent somatic cell hybrids carrying a single, intact, selectable human chromosome are valuable both for functional somatic cell genetic analysis and genome mapping procedures. Here, we describe the construction and detailed molecular cytogenetic characterization of a panel of 23 stable hybrids, representing all 22 human autosomes plus the X-chromosome. Individual normal human chromosomes have been tagged with a selectable fusion gene (Hytk) introduced into the chromosome in a small (4.2 kbp) retroviral vector. Use of the Hytk marker permits both positive and negative ("in-out") selection to be applied to the human chromosome in any mammalian cell background. The panel includes 18 new hybrids isolated by direct microcell transfer from normal human diploid fibroblasts into mouse A9 cells.


Sujet(s)
Cartographie chromosomique/méthodes , Test de complémentation/méthodes , Cellules hybrides , Adulte , Animaux , Diploïdie , Fibroblastes , Marqueurs génétiques , Humains , Caryotypage , Mâle , Souris , Phosphotransferases (Alcohol Group Acceptor)/génétique , Réaction de polymérisation en chaîne , Sites étiquetés par des séquences
9.
Toxicol Lett ; 67(1-3): 211-30, 1993 Apr.
Article de Anglais | MEDLINE | ID: mdl-8451762

RÉSUMÉ

Recent advances using somatic cell genetic approaches have provided a convincing body of evidence that the senescence of mammalian cells in culture is controlled by a small group of genes, one or more of which are functionally deleted in the process of immortalization. Microcell-mediated mono-chromosomal transfer methods should permit precise mapping of these genes to specific chromosomal regions. Cloning of senescence genes, using either conventional 'positional cloning' techniques or retroviral insertion mutagenesis, is now a realistic possibility. The leap in our understanding of the molecular genetic events driving the alternative cellular states of limited proliferative capacity and immortality, which such advances should precipitate, will finally permit the question of the role of cell immortalization in cancer to be addressed, and may open the door to the design of new modes of cancer therapy. In addition, the precise mechanism underlying the wide difference in transformability between human and rodent cells, which should also emerge from these investigations, is likely to make a significant contribution towards resolving the key issue of the relevance of rodent tumour induction assays in assessing the potential carcinogenicity of environmental chemicals.


Sujet(s)
Survie cellulaire/physiologie , Transformation cellulaire néoplasique , Vieillissement de la cellule/physiologie , Animaux , Lignée de cellules transformées , Vieillissement de la cellule/génétique , Clonage moléculaire , Cricetinae , Humains , Mesocricetus , Spécificité d'espèce
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE