Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Nat Genet ; 20(4): 337-43, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9843204

RESUMO

Leigh Syndrome (LS) is a severe neurological disorder characterized by bilaterally symmetrical necrotic lesions in subcortical brain regions that is commonly associated with systemic cytochrome c oxidase (COX) deficiency. COX deficiency is an autosomal recessive trait and most patients belong to a single genetic complementation group. DNA sequence analysis of the genes encoding the structural subunits of the COX complex has failed to identify a pathogenic mutation. Using microcell-mediated chromosome transfer, we mapped the gene defect in this disorder to chromosome 9q34 by complementation of the respiratory chain deficiency in patient fibroblasts. Analysis of a candidate gene (SURF1) of unknown function revealed several mutations, all of which predict a truncated protein. These data suggest a role for SURF1 in the biogenesis of the COX complex and define a new class of gene defects causing human neurodegenerative disease.


Assuntos
Complexo IV da Cadeia de Transporte de Elétrons/biossíntese , Doença de Leigh/genética , Mutação , Proteínas/genética , Sequência de Aminoácidos , Linhagem Celular , Mapeamento Cromossômico , Cromossomos Humanos Par 9 , DNA Complementar , Humanos , Hibridização in Situ Fluorescente , Proteínas de Membrana , Proteínas Mitocondriais , Dados de Sequência Molecular , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos
2.
J Natl Cancer Inst ; 91(1): 37-45, 1999 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-9890168

RESUMO

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.


Assuntos
Neoplasias da Mama/patologia , Carcinoma Intraductal não Infiltrante/patologia , Cromossomos Humanos Par 3/genética , Regulação Enzimológica da Expressão Gênica , Técnicas de Transferência de Genes , Inibidores do Crescimento/genética , Proteínas de Neoplasias/biossíntese , Proteínas Repressoras/genética , Telomerase/biossíntese , Adulto , Carcinoma Intraductal não Infiltrante/enzimologia , Divisão Celular/efeitos dos fármacos , Fusão Celular , Cromossomos/metabolismo , Cromossomos/ultraestrutura , Feminino , Inibidores do Crescimento/fisiologia , Humanos , Células Híbridas , Hibridização in Situ Fluorescente , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiologia , Reação em Cadeia da Polimerase , Proteínas Repressoras/fisiologia , Telomerase/genética , Telomerase/fisiologia , Telômero/metabolismo , Células Tumorais Cultivadas
3.
J Natl Cancer Inst ; 93(11): 865-72, 2001 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-11390536

RESUMO

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.


Assuntos
Cromossomos Humanos Par 6 , Papillomaviridae/genética , RNA , Telomerase/metabolismo , Neoplasias do Colo do Útero/genética , Divisão Celular , Linhagem Celular Transformada , Cromossomos Humanos Par 11 , Proteínas de Ligação a DNA , Feminino , Genes Reporter , Humanos , Células Híbridas , Queratinócitos , Repetições de Microssatélites , Reação em Cadeia da Polimerase , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Telomerase/antagonistas & inibidores , Telômero/genética , Telômero/ultraestrutura , Transfecção , Células Tumorais Cultivadas , beta-Galactosidase/genética
4.
Cancer Res ; 58(13): 2863-8, 1998 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-9661903

RESUMO

Interleukin 6 (IL-6) serves as a growth factor for mouse plasmacytomas. As a model for IL-6-mediated growth of plasmacytomas, we study IL-6-dependent B-cell hybridomas, which can be generated through fusion of B lymphocytes with a plasmacytoma cell line, e.g., SP2/0. In the present report, we have investigated the peculiar behavior of B-cell hybridomas with respect to IL-6 dependence. We demonstrate that although newly generated hybridomas are IL-6 dependent, many hybridomas lose this dependency at frequencies as high as 50%, shortly after fusion. We speculated that the loss of IL-6-dependent growth is due to the well-known chromosomal instability of B-cell hybridomas. Consequently, loss of IL-6 dependence is the result of loss of a specific chromosome(s). This model implies the existence of an "IL-6 dependency" gene, the loss of which makes hybridomas capable of proliferating in the absence of IL-6. Because SP2/0 is IL-6 independent, the IL-6-dependent phenotype of B-cell hybridomas, and hence the IL-6 dependency gene, must be derived from the B lymphocyte. We have tested this model by generating human/mouse B-cell hybridomas through fusion of human B lymphocytes with SP2/0. We then analyzed the human chromosome content of 10 IL-6-dependent and 14 IL-6-independent subclones. From that analysis we concluded that the presence of human chromosome 21 correlated with IL-6 dependence. This correlation was confirmed by microcell fusion experiments in which a single copy of chromosome 21 was introduced into IL-6-independent hybridomas, resulting in reconstitution of the IL-6-dependent phenotype. We therefore conclude that chromosome 21 carries an IL-6 dependency gene.


Assuntos
Cromossomos Humanos Par 21/fisiologia , Hibridomas , Interleucina-6/genética , Animais , Linfócitos B , Divisão Celular/genética , Cromossomos Humanos Par 21/genética , Feminino , Humanos , Hibridomas/citologia , Interleucina-6/fisiologia , Cariotipagem , Camundongos , Fenótipo
5.
Cancer Res ; 58(20): 4572-6, 1998 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-9788603

RESUMO

Genomic aberrations at the chromosome 16q arm are one of the most consistent abnormalities observed by loss of heterozygosity and comparative genomic hybridization analyses in human prostate cancer, suggesting that there are tumor suppressor or metastasis suppressor genes encoded by this chromosomal region. To functionally identify such suppressor genes, we have conducted microcell-mediated chromosome transfer to introduce human chromosome 16 into the highly metastatic Dunning rat prostatic cancer cell line, AT6.1. The metastatic ability of the resultant microcell hybrid clones was then tested in a standard spontaneous metastasis assay using SCID mice. When the microcell-mediated chromosome transfer hybrid cells containing whole human chromosome 16 were injected, the number of metastatic lesions in the lung was significantly reduced as much as 99% on average. Therefore, chromosome 16 has a strong activity to suppress the metastatic ability of AT6.1 cells while it did not affect the tumorigenesis and tumor growth rate. A PCR analysis of various microcell hybrid clones with sequence-tagged site markers indicates that the metastasis suppressor activity is located in the q24.2 region of chromosome 16. Our results are consistent with the previous finding that the region of human chromosome 16q has frequent loss of heterozygosity in prostate cancer patients and suggest that there is a metastasis suppressor gene in this region that may play an important role in the progression of prostate cancer.


Assuntos
Cromossomos Humanos Par 16 , Neoplasias Pulmonares/secundário , Neoplasias da Próstata/patologia , Animais , Caderinas/genética , Humanos , Perda de Heterozigosidade , Neoplasias Pulmonares/prevenção & controle , Masculino , Camundongos , Camundongos SCID , Neoplasias da Próstata/genética , Ratos , Transfecção , Células Tumorais Cultivadas
6.
Cancer Res ; 61(20): 7594-602, 2001 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-11606399

RESUMO

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.


Assuntos
Cromossomos Humanos Par 3/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Proto-Oncogênicas c-myc/fisiologia , RNA Mensageiro/antagonistas & inibidores , Telomerase/antagonistas & inibidores , Telomerase/metabolismo , Neoplasias da Mama/enzimologia , Neoplasias da Mama/genética , Ciclo Celular/genética , Diferenciação Celular/genética , Núcleo Celular/genética , Núcleo Celular/metabolismo , Proteínas de Ligação a DNA , Regulação para Baixo , Fibroblastos/enzimologia , Regulação Neoplásica da Expressão Gênica , Humanos , Proteínas Proto-Oncogênicas c-myc/genética , Splicing de RNA , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Neoplásico/genética , RNA Neoplásico/metabolismo , Telomerase/biossíntese , Telomerase/genética , Células Tumorais Cultivadas
7.
Cancer Res ; 59(3): 516-20, 1999 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-9973191

RESUMO

Losses of heterozygosity involving chromosomes 9 and 10 are frequent events in the development and progression of cutaneous malignant melanoma. To investigate whether specifically deleted chromosomal regions encode tumor suppressor genes (TSGs), we introduced normal chromosome 10 into the tumorigenic human metastatic melanoma cell line UACC-903 by microcell fusion. In addition, two chromosome 9 derivatives that were microdeleted in the region of the p16INK4A/p15INK4B locus were transferred to determine whether an additional melanoma TSG or TSGs reside on chromosome 9p, as indicated by previous melanoma allele loss studies. In comparison to parental cells, microcell hybrids generated with chromosomes 9 (microdeleted) and 10 displayed reduced anchorage-independent growth in soft agar and markedly reduced tumorigenicity in athymic (nu/nu) mice. These data define a TSG or TSGs that function independently of p15/p16 on chromosome 9 and provide evidence for a TSG (or TSGs) on chromosome 10 that may be important in melanoma development.


Assuntos
Proteínas de Ciclo Celular , Inibidor p16 de Quinase Dependente de Ciclina , Genes Supressores de Tumor , Melanoma/genética , Neoplasias Cutâneas/genética , Proteínas Supressoras de Tumor , Ágar , Animais , Proteínas de Transporte/genética , Divisão Celular/fisiologia , Deleção Cromossômica , Mapeamento Cromossômico , Cromossomos Humanos Par 10 , Cromossomos Humanos Par 9 , Clonagem Molecular , Inibidor de Quinase Dependente de Ciclina p15 , Feminino , Genes p16 , Humanos , Melanoma/patologia , Camundongos , Camundongos Nus , Células-Tronco Neoplásicas/patologia , Fenótipo , Neoplasias Cutâneas/patologia , Células Tumorais Cultivadas
8.
Cancer Res ; 59(9): 2038-40, 1999 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-10232582

RESUMO

Approximately 50% of immortal human keratinocyte lines show loss of heterozygosity of chromosome region 4q33-q34, and the reintroduction of chromosome 4 into one such line, BICR 6, causes proliferation arrest and features of replicative senescence. Recently, a candidate gene, mortality factor 4 (MORF4), was identified in this region and sequenced in 21 immortal keratinocyte lines. There were no mutations or deletions, and two of the seven lines that showed loss of heterozygosity at 4q33-q34 were heterozygous for MORF4 itself. Furthermore, the transfer of a chromosomal segment containing the entire MORF4 gene did not mimic the senescence effect of chromosome 4 in BICR 6. These results suggest that the inactivation of MORF4 is not required for human keratinocyte immortality.


Assuntos
Senescência Celular/genética , Queratinócitos/citologia , Fatores de Transcrição/fisiologia , Linhagem Celular Transformada , Cromossomos Humanos Par 4/genética , Fibroblastos/citologia , Genótipo , Humanos , Perda de Heterozigosidade , Reação em Cadeia da Polimerase , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Fatores de Transcrição/genética , Células Tumorais Cultivadas
9.
Oncogene ; 17(26): 3417-26, 1998 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-10030665

RESUMO

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.


Assuntos
Senescência Celular/fisiologia , Telomerase/metabolismo , Telômero/fisiologia , Animais , Antígenos Transformantes de Poliomavirus/genética , Divisão Celular , Linhagem Celular Transformada , Células Cultivadas , Cricetinae , Meios de Cultura Livres de Soro , Fatores de Tempo
10.
Oncogene ; 22(11): 1737-46, 2003 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-12642877

RESUMO

Normal human keratinocytes possess a finite replicative lifespan. Most advanced squamous cell carcinomas (SCCs), however, are immortal, a phenotype that is associated with p53 and INK4A dysfunction, high levels of telomerase and loss of heterozygosity (LOH) at several genetic loci, suggestive of the dysfunction of other mortality genes. We show here that human chromosome 6 specifically reduces the proliferation or viability of a human SCC line, BICR31, possessing LOH across the chromosome. This was determined by an 88% reduction in colony yield (P<0.001), following the reintroduction of an intact normal chromosome 6 by monochromosome transfer. Deletion analysis of immortal segregants using polymorphic markers revealed the loss of a 2.9 Mbp interval, centred on marker D6S1045 at 6q14.3-q15, in 6/19 segregants. Crucially, allelic losses of this region were not identified in control hybrids constructed between chromosome 6 and the BICR6 SCC cell line that is heterozygous for chromosome 6 and which showed no reduction in colony formation relative to the control chromosome transfers. This indicates that the minimally deleted region at D6S1045 is not the result of fragile sites, a recombination hot spot, or a feature of the monochromosome transfer technique. LOH of D6S1045 was found in 2/9 immortal SCC lines and was part of a minimally deleted region of line BICR19. Furthermore, allelic imbalance, consistent with LOH, was detected in 3/17 advanced SCCs of the tongue. These results suggest the existence of a suppressor of SCC immortality and tumour development at chromosome 6q14.3-q15, which is important to a subset of human SCCs.


Assuntos
Carcinoma de Células Escamosas/genética , Cromossomos Humanos Par 6 , Deleção de Genes , Carcinoma de Células Escamosas/mortalidade , Humanos , Perda de Heterozigosidade
11.
Clin Exp Metastasis ; 17(8): 663-8, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10919711

RESUMO

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


Assuntos
Neoplasias do Colo/genética , Proteínas do Citoesqueleto/genética , Proteínas de Ligação a DNA/deficiência , Inativação Gênica/fisiologia , Genes Supressores de Tumor/genética , Alelos , Animais , Pareamento Incorreto de Bases , Fusão Celular , Cromossomos Humanos Par 2/genética , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Proteínas do Citoesqueleto/biossíntese , Reparo do DNA , Proteínas de Ligação a DNA/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Camundongos , Fenótipo , Poliploidia , Transfecção , Células Tumorais Cultivadas , alfa Catenina
12.
Toxicol Lett ; 67(1-3): 211-30, 1993 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-8451762

RESUMO

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.


Assuntos
Sobrevivência Celular/fisiologia , Transformação Celular Neoplásica , Senescência Celular/fisiologia , Animais , Linhagem Celular Transformada , Senescência Celular/genética , Clonagem Molecular , Cricetinae , Humanos , Mesocricetus , Especificidade da Espécie
13.
Mutat Res ; 364(2): 91-102, 1996 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-8879275

RESUMO

X-ray sensitive Chinese hamster V79 cells mutants, V-C4, V-E5 and V-G8, show an abnormal response to X-ray-induced DNA damage. Like ataxia telangiectasia (AT) cells, they display increased cell killing, chromosomal instability and a diminished inhibition of DNA synthesis following ionizing radiation. To localize the defective hamster gene (XRCC8) on the human genome, human chromosomes were introduced into the AT-like hamster mutants, by microcell mediated chromosome transfer. Although, none of the human chromosomes corrected the defect in these mutants, the defect was corrected by a single mouse chromosome, derived from the A9 microcell donor cell line. In four independent X-ray-resistant microcell hybrid clones of V-E5, the presence of the mouse chromosome was determined by fluorescent in situ hybridization, using a mouse cot-1 probe. By PCR analysis with primers specific for different mouse chromosomes and Southern blot analysis with the mouse Ldlr probe, the mouse chromosome 9, was identified in all four X-ray-resistant hybrid clones. Segregation of the mouse chromosome 9 from these hamster-mouse microcell hybrids led to the loss of the regained X-ray-resistance, confirming that mouse chromosome 9 is responsible for complementation of the defect in V-E5 cells. The assignment of the mouse homolog of the ATM gene to mouse chromosome 9, and the presence of this mouse chromosome only in the radioresistant hamster cell hybrids suggest that the hamster AT-like mutant are homologous to AT, although they are not complemented by hamster chromosome 11.


Assuntos
Ataxia Telangiectasia/genética , Cricetinae/genética , Mutação , Tolerância a Radiação/genética , Animais , Sobrevivência Celular , Células Cultivadas , Aberrações Cromossômicas , Cromossomos , Relação Dose-Resposta à Radiação , Raios gama , Teste de Complementação Genética , Humanos , Camundongos , Especificidade da Espécie , Raios X
14.
Mol Carcinog ; 12(4): 213-24, 1995 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-7727043

RESUMO

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.


Assuntos
Deleção Cromossômica , Clonagem Molecular , Deleção de Genes , Marcadores Genéticos , Células 3T3 , 7,8-Di-Hidro-7,8-Di-Hidroxibenzo(a)pireno 9,10-óxido/toxicidade , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Relação Dose-Resposta à Radiação , Genes Supressores de Tumor , Variação Genética , Humanos , Camundongos , Nêutrons , Plasmídeos , Reação em Cadeia da Polimerase , Provírus , Retroviridae , Transfecção
15.
Carcinogenesis ; 16(2): 193-204, 1995 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-7859348

RESUMO

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.


Assuntos
Carcinógenos/toxicidade , Transformação Celular Neoplásica , Fibroblastos/efeitos dos fármacos , Fibroblastos/efeitos da radiação , Pele/efeitos dos fármacos , Pele/efeitos da radiação , Animais , Transformação Celular Neoplásica/genética , Cromossomos/efeitos dos fármacos , Cromossomos/efeitos da radiação , Células Clonais , Cricetinae , Cricetulus , Relação Dose-Resposta a Droga , Relação Dose-Resposta à Radiação , Fibroblastos/citologia , Técnicas de Transferência de Genes , Genes/efeitos dos fármacos , Genes/efeitos da radiação , Humanos , Cariotipagem , Mesocricetus , Camundongos , Fenótipo , Ratos , Pele/citologia , Transformação Genética
16.
Ann Hum Genet ; 67(Pt 6): 543-56, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14641242

RESUMO

Association studies for disease susceptibility genes rely on the high density of SNPs within candidate genes. However, the linkage disequilibrium between SNPs imply that not all SNPs identified in the candidate region need be genotyped. Here we develop several approaches to SNP subset selection, which can substantially reduce the number of SNPs to be genotyped in an association study. We apply clustering algorithms to pairwise linkage disequilibrium measures, with SNP subsets determined for different cut-off values of Delta using nearest and furthest neighbour clusters. Alternatively, SNP subsets may be determined by the proportion of haplotypes they identify. We also show how power calculations, based on the average power to identify a SNP as the disease susceptibility mutation using haplotype-based or logistic regression based statistical analyses, can be used to choose SNP subsets. All these methods provide a ranking method for subsets of a specific size, but do not provide criteria for overall choice of SNP subset size. We develop such criteria by incorporating power calculations into a decision analysis, where the choice of SNP subset size depends on the genotyping costs and the perceived benefits of identifying association. These methods are illustrated using eleven SNPs in the MMP2 gene.


Assuntos
Desequilíbrio de Ligação/genética , Modelos Genéticos , Polimorfismo de Nucleotídeo Único/genética , Projetos de Pesquisa , Análise por Conglomerados , Teoria da Decisão , Frequência do Gene , Genótipo , Haplótipos/genética , Humanos , Metaloproteinase 2 da Matriz/genética , Análise de Regressão , Tamanho da Amostra
17.
EMBO J ; 20(11): 2867-74, 2001 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-11387219

RESUMO

Human ribosomal gene repeats are distributed among five nucleolar organizer regions (NORs) on the p arms of acrocentric chromosomes. On exit from mitosis, nucleoli form around individual active NORs. As cells progress through the cycle, these mini-nucleoli fuse to form large nucleoli incorporating multiple NORs. It is generally assumed that nucleolar incorporation of individual NORs is dependent on ribosomal gene transcription. To test this assumption, we determined the nuclear location of individual human acrocentric chromosomes, and their associated NORs, in mouse> human cell hybrids. Human ribosomal genes are transcriptionally silent in this context. Combined immunofluorescence and in situ hybridization (immuno-FISH) on three-dimensional preserved nuclei showed that human acrocentric chromosomes associate with hybrid cell nucleoli. Analysis of purified nucleoli demonstrated that human and mouse NORs are equally likely to be within a hybrid cell nucleolus. This is supported further by the observation that murine upstream binding factor can associate with human NORs. Incorporation of silent NORs into mature nucleoli raises interesting issues concerning the maintenance of the activity status of individual NORs.


Assuntos
Cromossomos Humanos/genética , DNA Ribossômico/genética , Inativação Gênica , Região Organizadora do Nucléolo/genética , RNA Ribossômico 28S/genética , Transcrição Gênica , Animais , Linhagem Celular , Núcleo Celular/genética , Núcleo Celular/ultraestrutura , Cromossomos Humanos/ultraestrutura , Imunofluorescência , Células HeLa , Humanos , Células Híbridas , Hibridização in Situ Fluorescente , Metáfase , Camundongos , Região Organizadora do Nucléolo/ultraestrutura , Reação em Cadeia da Polimerase
18.
Carcinogenesis ; 17(8): 1567-75, 1996 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8761411

RESUMO

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.


Assuntos
Proteínas de Transporte/genética , Cromossomos Humanos Par 9 , Genes Supressores de Tumor , Animais , Sequência de Bases , Divisão Celular/genética , Linhagem Celular Transformada , Inibidor p16 de Quinase Dependente de Ciclina , Primers do DNA , Marcadores Genéticos , Humanos , Células Híbridas , Melanoma/genética , Dados de Sequência Molecular , Transfecção , Células Tumorais Cultivadas
19.
Cytogenet Cell Genet ; 71(1): 68-76, 1995.
Artigo em Inglês | MEDLINE | ID: mdl-7606932

RESUMO

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.


Assuntos
Mapeamento Cromossômico/métodos , Teste de Complementação Genética/métodos , Células Híbridas , Adulto , Animais , Diploide , Fibroblastos , Marcadores Genéticos , Humanos , Cariotipagem , Masculino , Camundongos , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Reação em Cadeia da Polimerase , Sitios de Sequências Rotuladas
20.
Eur J Immunogenet ; 30(3): 187-90, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12786995

RESUMO

The immunoglobulin superfamily 6 gene (IGSF6) on chromosome 16p11-p12 has been investigated as a positional and functional candidate for inflammatory bowel disease (IBD) susceptibility. Screening of the six exons of IGSF6 for single nucleotide polymorphisms (SNPs) detected four novel SNPs, and validated three of six SNPs listed in the international SNP database (dbSNP). The seven SNPs in IGSF6 formed five distinct linkage disequilibrium groups. There was no evidence for association of the common SNPs with disease in a large cohort of patients with IBD. The novel SNPs and the linkage disequilibrium map will be a useful resource for the analysis of IGSF6 in other immune disorders.


Assuntos
Antígenos CD8/genética , Predisposição Genética para Doença , Variação Genética , Imunoglobulinas/genética , Doenças Inflamatórias Intestinais/genética , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Transporte/genética , Estudos de Casos e Controles , Colite Ulcerativa/genética , Doença de Crohn/genética , Humanos , Desequilíbrio de Ligação , Proteína Adaptadora de Sinalização NOD2 , Linhagem , Polimorfismo de Nucleotídeo Único
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA