Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Nat Genet ; 7(1): 29-33, 1994 May.
Artículo en Inglés | MEDLINE | ID: mdl-8075635

RESUMEN

Chromosome sorting from hybrid cells offers enormous advantages for gene mapping and cloning, but purification of most chromosomes has been largely hindered by their similarity in size to other chromosomes. We have developed a novel cell line and strategy that allows simple, mass purification of mammalian chromosomes, permitting significant target genome enrichment. This strategy takes advantage of the small number of giant chromosomes (1,2,X) of the female Indian muntjac, a barking deer, avoiding the problem of size similarity. We introduced human chromosomes into a cell line derived from a muntjac and purified them to homogeneity using a relatively simple technique. This strategy should facilitate the isolation of chromosomes from species other than human for which hybrid cells are not available currently.


Asunto(s)
Fraccionamiento Celular/métodos , Centrifugación por Gradiente de Densidad , Cromosomas Humanos , Ciervos/genética , Citometría de Flujo , Células Híbridas/ultraestructura , Animales , Línea Celular Transformada , Transformación Celular Viral , Cromosomas Humanos Par 11 , ADN/análisis , Femenino , Fibroblastos/citología , Humanos , Hibridación Fluorescente in Situ , Virus 40 de los Simios
2.
Science ; 272(5261): 557-60, 1996 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-8614807

RESUMEN

Deficiencies in mismatch repair have been linked to a common cancer predisposition syndrome in humans, hereditary nonpolyposis colorectal cancer (HNPCC), and a subset of sporadic cancers. Here, several mismatch repair-deficient tumor cell lines and HNPCC-derived lymphoblastoid cell lines were found to be deficient in an additional DNA repair process termed transcription-coupled repair (TCR). The TCR defect was corrected in a mutant cell line whose mismatch repair deficiency had been corrected by chromosome transfer. Thus, the connection between excision repair and mismatch repair previously described in Escherichia coli extends to humans. These results imply that deficiencies in TCR and exposure to carcinogens present in the environment may contribute to the etiology of tumors associated with genetic defects in mismatch repair.


Asunto(s)
Adenosina Trifosfatasas , Neoplasias Colorrectales Hereditarias sin Poliposis/genética , Enzimas Reparadoras del ADN , Reparación del ADN , Proteínas de Unión al ADN , Mutación , Neoplasias/genética , Transcripción Genética , Daño del ADN , Humanos , Linfocitos/citología , Endonucleasa PMS2 de Reparación del Emparejamiento Incorrecto , Proteína 2 Homóloga a MutS , Proteínas de Neoplasias/genética , Proteínas Proto-Oncogénicas/genética , Dímeros de Pirimidina/metabolismo , Tetrahidrofolato Deshidrogenasa/genética , Células Tumorales Cultivadas , Rayos Ultravioleta
3.
Science ; 247(4943): 707-10, 1990 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-2300822

RESUMEN

The control of cellular senescence by specific human chromosomes was examined in interspecies cell hybrids between diploid human fibroblasts and an immortal, Syrian hamster cell line. Most such hybrids exhibited a limited life span comparable to that of the human fibroblasts, indicating that cellular senescence is dominant in these hybrids. Karyotypic analyses of the hybrid clones that did not senesce revealed that all these clones had lost both copies of human chromosome 1, whereas all other human chromosomes were observed in at least some of the immortal hybrids. The application of selective pressure for retention of human chromosome 1 to the cell hybrids resulted in an increased percentage of hybrids that senesced. Further, the introduction of a single copy of human chromosome 1 to the hamster cells by microcell fusion caused typical signs of cellular senescence. Transfer of chromosome 11 had no effect on the growth of the cells. These findings indicate that human chromosome 1 may participate in the control of cellular senescence and further support a genetic basis for cellular senescence.


Asunto(s)
Supervivencia Celular/genética , Cromosomas Humanos Par 1 , Fibroblastos/citología , Células Híbridas/citología , Animales , Línea Celular , Mapeo Cromosómico , Células Clonales , Cricetinae , Diploidia , Humanos , Hipoxantina Fosforribosiltransferasa/genética , Cariotipificación , Ratones , Ploidias , Transfección , Translocación Genética , Cromosoma X
4.
Science ; 260(5106): 361-4, 1993 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-8469989

RESUMEN

A fundamental problem in the identification and isolation of tumor suppressor and other growth-inhibiting genes is the loss of power of genetic complementation at the subchromosomal level. A direct genetic strategy was developed to isolate subchromosomal transferable fragments (STFs) from any chromosome, each containing a selectable marker within the human DNA, that could be transferred to any mammalian cell. As a test of the method, several overlapping STFs from 11p15 were shown to cause in vitro growth arrest of rhabdomyosarcoma cells. This activity mapped between the beta-globin and insulin genes.


Asunto(s)
Cromosomas Humanos Par 11 , ADN/genética , Genes Supresores de Tumor , Técnicas Genéticas , Rabdomiosarcoma/patología , Animales , Secuencia de Bases , Células CHO , División Celular , Línea Celular , Cricetinae , Marcadores Genéticos , Globinas/genética , Humanos , Insulina/genética , Ratones , Datos de Secuencia Molecular , Células Tumorales Cultivadas
5.
Science ; 250(4983): 994-7, 1990 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-2173146

RESUMEN

Wilms tumor is an embryonal kidney tumor involving complex pathology and genetics. The Wilms tumor locus on chromosome 11p13 is defined by the region of overlap of constitutional and tumor-associated deletions. Chromosome walking and yeast artificial chromosome (YAC) cloning were used to clone and map 850 kilobases of DNA. Nine CpG islands, constituting a "CpG island archipelago," were identified, including three islands that were not apparent by conventional pulsed-field mapping, and thus were at least partially methylated. Three distinct transcriptional units were found closely associated with a CpG island within the boundaries of a homozygous DNA deletion in a Wilms tumor.


Asunto(s)
Mapeo Cromosómico , Cromosomas Humanos Par 11 , Fosfatos de Dinucleósidos , Genes del Tumor de Wilms/genética , Tumor de Wilms/genética , Paseo de Cromosoma , Sondas de ADN , Humanos , Transcripción Genética
6.
J Clin Invest ; 98(1): 199-206, 1996 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-8690794

RESUMEN

The phenomenon of alkylation tolerance has been observed in cells that are deficient in some component of the DNA mismatch repair (MMR) system. An alkylation-induced cell cycle arrest had been reported previously in one MMR-proficient cell line, whereas a MMR-defective clone derived from this line escapes from this arrest. We examined human cancer cell lines to determine if the cell cycle arrest were dependent upon the MMR system. Growth characteristics and cell cycle analysis after MNNG treatment were ascertained in seven MMR-deficient and proficient cell lines, with and without confirmed mutations in hMLH1 or hMSH2 by an in vitro transcription/translation assay. MMR-proficient cells underwent growth arrest in the G2 phase of the cell cycle after the first S phase, whereas MMR-deficient cells escaped an initial G2 delay and resumed a normal growth pattern. In the HCT116 line corrected for defective MMR by chromosome 3 transfer, the G2 phase arrest lasted more than five days. In another MMR-proficient colon cancer cell line, SW480, cell death occurred five days after MNNG treatment. A competent MMR system appears to be necessary for G2 arrest or cell death after alkylation damage, and this cell cycle checkpoint may allow the cell to repair damaged DNA, or prevent the replication of mutated DNA by prohibiting clonal expansion.


Asunto(s)
Daño del ADN , Reparación del ADN , Proteínas de Unión al ADN , Fase G2/efectos de los fármacos , Metilnitronitrosoguanidina/farmacología , Neoplasias/genética , Alquilantes/farmacología , Carcinoma , Neoplasias del Colon , Femenino , Humanos , Modelos Genéticos , Proteína 2 Homóloga a MutS , Neoplasias Ováricas , Proteínas Proto-Oncogénicas/genética , ARN Mensajero/análisis , Eliminación de Secuencia , Células Madre , Células Tumorales Cultivadas
7.
Cancer Res ; 48(6): 1623-32, 1988 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-2449958

RESUMEN

It has been shown previously that normal Syrian hamster embryo cells are neoplastically transformed by transfection with two cooperating oncogenes, v-myc plus v-Ha-ras. Karyotypic analyses of the cells from the tumors revealed a nonrandom chromosome change, monosomy of chromosome 15. In order to clarify the role of chromosome loss in these tumor cells with defined oncogene alterations, molecular and cytogenetic studies were performed on hybrids between normal Syrian hamster embryo cells and ras/myc tumor cells. Following fusion of the tumor cells with the normal cells which are not immortal, the majority of the cell hybrids senesced after less than or equal to 20 population doublings indicating that immortality was recessive. Some of the hybrids escaped senescence and grew indefinitely. These immortal hybrid cells retained the expected numbers of chromosome 15 indicating that escape from senescence did not involve loss of this chromosome. The tumorigenicity and anchorage-independent growth of the nonsenescent hybrids were still suppressed significantly. In these suppressed hybrid cells, RNAs complementary to the v-Ha-ras and v-myc oncogenes were expressed. Furthermore, radioimmune precipitation with a monoclonal antibody to p21ras of [35S]methionine-labeled cell extracts followed by polyacrylamide gel electrophoresis/sodium dodecyl sulfate electrophoresis showed that the suppressed hybrid cells contained high levels of the mutated ras protein. These results indicate that tumorigenicity is suppressed in the hybrids even though the oncogenes are expressed. When the hybrid cells were passaged, anchorage-independent variants appeared in the cultures. At this time, morphological changes occurred in the cultures and the cells were tumorigenic. Karyotypic analyses of the transformed segregants versus the parental hybrid cells revealed a nonrandom loss of one copy of chromosome 15 in the transformed segregants. No other nonrandom chromosome change was observed. These results suggest that the loss of chromosome 15 results in the loss of a cellular tumor suppressor gene which effects a phenotypic change necessary for expression of neoplastic transformation. In addition, the cellular factors responsible for the senescence of the hybrids may provide another mechanism involved in suppressing tumorigenicity.


Asunto(s)
Transformación Celular Neoplásica , Aberraciones Cromosómicas , Oncogenes , Animales , Línea Celular , Supervivencia Celular , Cricetinae , Cariotipificación , Mesocricetus , Metafase , Hibridación de Ácido Nucleico , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas p21(ras) , ARN/análisis , Transfección
8.
Cancer Res ; 54(16): 4308-12, 1994 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-8044777

RESUMEN

The human colon tumor cell line HCT 116 is known to have a homozygous mutation in the mismatch repair gene hMLH1 on human chromosome 3, to exhibit microsatellite instability, and to be defective in mismatch repair. In order to determine whether the introduction of a normal copy of hMLH1 gene restores mismatch repair activity and corrects microsatellite instability, a single human chromosome 3 from normal fibroblasts was transferred to HCT 116 cells via microcell fusion. As a control, human chromosome 2 was also transferred to HCT 116 cells. Two HCT 116 microcell hybrid clones that received a single copy of chromosome 2 (HCT 116 + ch2) and two that received a single copy of chromosome 3 (HCT 116 + ch3) were isolated and characterized. A G-G mismatch in M13-derived heteroduplex DNA was efficiently repaired in cell extracts from HCT 116 + ch3 cells, but not in those of parent HCT 116 cells or HCT 116 + ch2 cells. Microsatellite alterations at the D5S107 locus containing CA repeats were seen in 8 of 80 subclones from HCT 116 cells, and in 13 of 150 subclones from HCT 116 + ch2 cells. In contrast, none of the 225 subclones derived from mismatch repair-proficient HCT 116 + ch3 cells showed alterations in the microsatellite at the same locus. The effect of introducing chromosome 3 on the sensitivity of HCT 116 cells to N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) was examined, since enhanced tolerance to MNNG is accompanied by loss of mismatch repair activity in several cell lines. Within 3 days after treatment with 5 microM MNNG, HCT 116 + ch3 cells became morphologically flat and stopped growing. Their colony-forming ability, determined 10 days after treatment, was reduced 200-fold when compared to MNNG-treated parental HCT 116 and HCT 116 + ch2 cells. These results support the hypothesis that mutations in both alleles of the hMLH1 gene are necessary for the manifestation of defective mismatch repair and microsatellite instability and for enhanced MNNG tolerance. The results also suggest that the mismatch repair system contributes to the process that causes growth arrest in response to DNA damage by alkylating agents.


Asunto(s)
Cromosomas Humanos Par 3/fisiología , Neoplasias del Colon/genética , Reparación del ADN/genética , ADN Satélite/genética , Metilnitronitrosoguanidina/farmacología , Mutación Puntual/genética , Secuencia de Bases , Bandeo Cromosómico , Cromosomas Humanos Par 2 , Tolerancia a Medicamentos , Humanos , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Transfección , Células Tumorales Cultivadas , Ensayo de Tumor de Célula Madre
9.
Cancer Res ; 56(13): 3087-90, 1996 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-8674066

RESUMEN

Selection of cells for resistance to cisplatin, a well-recognized mutagen, could result in mutations in genes involved in DNA mismatch repair and thereby to resistance to DNA-alkylating agents. Parental cells of the human ovarian adenocarcinoma cell line 2008 expressed hMLH1 when analyzed with immunoblot. One subline selected for resistance to cisplatin (2008/A) expressed no hMLH1, whereas another (2008/C13*5.25) expressed parental levels. Microsatellite instability was readily demonstrated in 2008/A cells but not in 2008 and in 2008/C13*5.25 cells. In addition, the 2008/A cells were 2-fold resistant to methyl-nitro-nitrosoguanidine and had a 65-fold elevated mutation rate at the HPRT locus as compared to 2008 cells, both of which are consistent with the loss of DNA mismatch repair in these cells. To determine whether the loss of DNA mismatch repair itself contributes to cisplatin resistance, studies were carried out in isogenic pairs of cell lines proficient or defective in this function. HCT116, a human colon cancer cell line deficient in hMLH1 function, was 2-fold resistant to cisplatin when compared to a subline complemented with chromosome 3 and expressing hMLH1. Similarly, the human endometrial cancer cell line HEC59, which expresses no hMSH2, was 2-fold resistant to cisplatin when compared to a subline complemented with chromosome 2 that expresses hMSH2. Therefore, the selection of cells for resistance to cisplatin can result in the loss of DNA mismatch repair, and loss of DNA mismatch repair in turn contributes to resistance to cisplatin.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/genética , Cisplatino/farmacología , Reparación del ADN , Proteínas de Unión al ADN , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Proteínas Adaptadoras Transductoras de Señales , Secuencia de Bases , Carcinógenos , Proteínas Portadoras , ADN de Neoplasias/genética , Resistencia a Antineoplásicos , Femenino , Humanos , Hipoxantina Fosforribosiltransferasa/genética , Immunoblotting , Metilnitronitrosoguanidina , Datos de Secuencia Molecular , Homólogo 1 de la Proteína MutL , Proteína 2 Homóloga a MutS , Mutación , Proteínas de Neoplasias/genética , Proteínas Nucleares , Proteínas Proto-Oncogénicas/metabolismo , Células Tumorales Cultivadas
10.
Cancer Res ; 57(18): 3949-55, 1997 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-9307278

RESUMEN

The human DNA mismatch repair genes hMSH2 and hMSH6 encode the proteins that, together, bind to mismatches to initiate repair of replication errors. Human tumor cells containing mutations in these genes have strongly elevated mutation rates in selectable genes and at microsatellite loci, although mutations in these genes cause somewhat different mutator phenotypes. These cells are also resistant to killing by certain drugs and are defective in mismatch repair. Because the elevated mutation rates in these cells may lead to mutations in additional genes that are causally related to the other defects, here we attempt to establish a cause-effect relationship between the hMSH2 and hMSH6 gene mutations and the observed phenotypes. The endometrial tumor cell line HEC59 contains mutations in both alleles of hMSH2. The colon tumor cell line HCT15 contains mutations in hMSH6 and also has a sequence change in a conserved region of the coding sequence for DNA polymerase delta, a replicative DNA polymerase. We introduced human chromosome 2 containing the wild-type hMSH2 and hMSH6 genes into HEC59 and HCT15 cells. Introduction of chromosome 2 to HEC59 cells restored microsatellite stability, sensitivity to N-methyl-N'-nitro-N-nitrosoguanidine treatment, and mismatch repair activity. Transfer of chromosome 2 to HCT15 cells also reduced the mutation rate at the HPRT locus and restored sensitivity to N-methyl-N'-nitro-N-nitrosoguanidine treatment and mismatch repair activity. The results demonstrate that the observed defects are causally related to mutations in genes on chromosome 2, probably hMSH2 or hMSH6, but are not related to sequence changes in other genes, including the gene encoding DNA polymerase delta.


Asunto(s)
Cromosomas Humanos Par 2 , Reparación del ADN , Proteínas de Unión al ADN/fisiología , Mutagénesis , Proteínas Proto-Oncogénicas/fisiología , Fusión Celular , Resistencia a Antineoplásicos , Humanos , Metilnitronitrosoguanidina/farmacología , Repeticiones de Microsatélite , Proteína 2 Homóloga a MutS , Eliminación de Secuencia , Células Tumorales Cultivadas
11.
Cancer Res ; 55(17): 3721-5, 1995 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-7641183

RESUMEN

The human colon tumor cell line HCT116 is deficient in wild-type hMLH1, is defective in mismatch repair (MMR), exhibits microsatellite instability, and is tolerant to N-methyl-N'-nitro-N-nitrosoguanidine (MNNG). Transferring a normal copy of hMLH1 on chromosome 3 into the cell line restores MMR activity, stabilizes microsatellite loci, and increases the sensitivity of the cell to MNNG. Previous studies in other cell lines tolerant to alkylating agents such as MNNG or N-methylnitrosourea have shown cross-tolerance to 6-thioguanine (6TG), leading to a hypothesis that tolerance to MNNG or 6TG may be the result of MMR deficiency. To test this hypothesis, we studied the effects of 6TG on the MNNG-tolerant, MMR-deficient HCT116 cell line and its MNNG-sensitive, MMR-proficient, MNNG-tolerant, and MMR-deficient derivatives. Continuous exposure to low doses of 6TG (0.31-1.25 micrograms/ml) had no apparent effect on colony-forming ability (CFA) in MNNG-tolerant, MMR-deficient cells, whereas MNNG-sensitive, MMR-proficient cells exhibited a dose-dependent decrease in CFA. Growth kinetics and cell cycle analysis revealed that the growth of 6TG-treated HCT116 + chr3 cells was arrested at G2 after exposure to low dose of 6TG. In contrast, the same exposure to 6TG did not induce G2 arrest but rather a G1 delay in HCT116 and HCT116 + chr2. To obtain further evidence for the role of MMR on 6TG and MNNG toxicity, we isolated an MNNG-resistant revertant clone, M2, from the MNNG-sensitive, MMR-proficient HCT116 + chr3 cell line and characterized the MMR activity, hMLH1 status, and 6TG response. The results showed that M2 cells lost MMR activity as well as the previously introduced normal hMLH1 gene. Restoration of the CFA of M2 and an absence of G2 arrest were observed after treatment with low doses of 6TG. These results suggest that the mismatch repair system interacts with the G2 checkpoint in response to 6TG or MNNG-induced DNA lesions. The results further suggest that any agent that induces DNA mispairs will cause G2 arrest in MMR-proficient cells but not in MMR-deficient cells.


Asunto(s)
Neoplasias Colorrectales Hereditarias sin Poliposis/genética , Reparación del ADN , Fase G2/efectos de los fármacos , Metilnitronitrosoguanidina/farmacología , Tioguanina/farmacología , Azaguanina/farmacología , Secuencia de Bases , División Celular/efectos de los fármacos , División Celular/genética , Neoplasias Colorrectales Hereditarias sin Poliposis/patología , Sondas de ADN , ADN de Neoplasias/efectos de los fármacos , ADN de Neoplasias/metabolismo , Resistencia a Medicamentos , Fase G2/genética , Humanos , Datos de Secuencia Molecular , Tioguanina/metabolismo , Células Tumorales Cultivadas , Ensayo de Tumor de Célula Madre
12.
Mutat Res ; 235(2): 59-63, 1990 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-2155385

RESUMEN

In order to identify the human chromosome which carries a mutated gene in cells from a patient with the hereditary disorder ataxia telangiectasia belonging to complementation group D (AT-D), we performed chromosome transfer experiments via microcell fusion. A single, pSV2neo-tagged chromosome, either 11 or 12, derived from normal human fibroblasts was introduced into AT-D cells by microcell fusion, and clones which were resistant to the antibiotic G418 were isolated. All 3 hybrid clones containing an additional copy number of chromosome 11 showed a restoration of the resistance of wild-type cells to killing by X-irradiation, whereas all 3 hybrid clones containing an additional copy number of chromosome 12 remained hyper-radiosensitive, like the parental AT cells. The results indicate that a defective gene of AT-D cells is also located on chromosome 11, since a genetic linkage analysis has previously suggested that a defective gene of its complementation group A is located on this chromosome.


Asunto(s)
Ataxia Telangiectasia/genética , Cromosomas Humanos Par 11 , Tolerancia a Radiación/genética , Ataxia Telangiectasia/inmunología , Línea Celular Transformada , Transformación Celular Viral , Cromosomas Humanos Par 12 , Humanos , Células Híbridas , Técnicas de Transferencia Nuclear , Virus 40 de los Simios
13.
Mutat Res ; 402(1-2): 15-22, 1998 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-9675233

RESUMEN

The study of the multiple functions of mismatch repair genes in humans is being facilitated by the use of human tumor cell lines carrying defined MMR gene mutations. Such cell lines have elevated spontaneous mutation rates and may accumulate mutations in other genes, some of which could be causally related to the phenotypes of these cells. One approach to establish a cause-effect relationship between a MMR gene defect and a phenotype is to determine if that phenotype is reversed when a normal chromosome carrying a wild-type MMR gene is introduced by microcell fusion. This approach has the advantage of presenting the gene in its natural chromosomal environment with normal regulatory controls and at a reasonable dosage. The approach also limits candidate genes to only those encoded by the introduced chromosome and not elsewhere in the genome. Here we review studies demonstrating that hMSH2, hMSH3, hMSH6 and hMLH1 gene defects can each be complemented by transferring human chromosome 2, 5, 2 or 3, respectively. These transfers restore MMR activity, sensitivity to killing by MNNG, stability to microsatellite sequences and low spontaneous HPRT gene mutation rates.


Asunto(s)
Cromosomas Humanos , Reparación del ADN/genética , Prueba de Complementación Genética , Ácidos Nucleicos Heterodúplex , Fusión Celular , Humanos , Células Híbridas , Mutación
14.
Int J Cancer ; 20(4): 572-80, 1977 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-914396

RESUMEN

A simple method for microinjecting isolated chromosomes into a single living cell under an inverted microscope has been developed. Of the 368 injected cells, 85 were able to form a colony and could be cloned. Clones of Chinese hamster V79 cells microinjected with chromosomes isolated from murine D56 cells [V79 (D56) cells] were tested for tumorigenicity in immunodeficient nude mice and for colony-forming ability in soft agar. Untreated recipient V79 cells were highly tumorigenic and had a high colony-forming ability in soft agar. In contrast, two out of the 21 microinjected clones tested were non-tumorigenic in nude mice and had only weak colony-forming ability in soft agar. The chromosome banding pattern was analyzed in microinjected clones and tumors derived from cells of these clones. In cells of the two non-tumorigenic clones, a telocentric chhromosome 1 (t1) was specifically involved in translocations with other chromosomes or chromosome fragments. In all tumor cells obtained from nude mice, a supernumerary piece or a whole biarmed chromosome 14 (b14) was specifically found. The results suggest that the t1 chromosome bears the gene which controls in vitro transformation and that the additional genetic change, i.e. the extra piece of b14 chromosome, was required for tumor formation in vivo.


Asunto(s)
Transformación Celular Viral , Cromosomas , Neoplasias Experimentales/genética , Animales , División Celular , Supervivencia Celular , Células Cultivadas , Células Clonales , Cricetinae , Metafase , Métodos , Ratones , Ratones Desnudos , Microinyecciones/instrumentación , Virus de la Leucemia Murina de Moloney , Translocación Genética
15.
Proc Natl Acad Sci U S A ; 83(16): 5992-6, 1986 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-3461473

RESUMEN

Cell hybrids between normal, early-passage Syrian hamster embryo cells and a highly tumorigenic, chemically transformed hamster cell line, BP6T, were formed, selected, and analyzed. Tumorigenicity and anchorage-independent growth were suppressed in the hybrid cells compared to the tumorigenic BP6T cells. These two phenotypes segregated coordinately in these cells. To determine at what stage in the neoplastic process this tumor-suppressive function was lost, two chemically induced immortal cell lines were examined at different passages for the ability to suppress the tumorigenic phenotype of BP6T cells following hybridization. Hybrids of BP6T cells with the immortal, nontumorigenic cell lines at early passages were suppressed for tumorigenicity and anchorage-independent growth. This tumor-suppressive ability was reduced in the same cells at later passages and in some cases nearly completely lost, prior to the neoplastic transformation of the immortal cell lines. Subclones of the cell lines were heterogeneous in their ability to suppress tumorigenicity in cell hybrids; some clones retained the tumor-suppressive ability and others lost this function. The susceptibility to neoplastic transformation of these cells following DNA transfection with the viral ras oncogene or BP6T DNA inversely correlated with the tumor-suppressive ability of the cells. These results suggest that chemically induced neoplastic progression of Syrian hamster embryo cells involves at least three steps: induction of immortality, activation of a transforming oncogene, and loss of a tumor-suppressive function.


Asunto(s)
Transformación Celular Neoplásica , Animales , Adhesión Celular , División Celular , Línea Celular , Células Cultivadas , Cricetinae , ADN de Neoplasias/aislamiento & purificación , Embrión de Mamíferos , Células Híbridas/citología , Mesocricetus , Oncogenes
16.
Jpn J Cancer Res ; 80(2): 122-5, 1989 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-2498246

RESUMEN

Cell hybrids between hypoxanthine guanine phosphoribosyl transferase (HGPRT)-deficient mouse cell lines (A9 or RAG) and each of 12 different human fibroblasts (GM cells) containing various X/autosome translocations were formed, selected and isolated. Several human chromosomes including an X/autosome translocation carrying HGPRT locus were found in these hybrid cells. To construct A9 cell clones that contain a single X/autosome translocation, micro-cell fusion was undertaken to transfer these chromosomes from the hybrids to A9 cells. Karyotype analysis revealed that most of the resulting micro-cell hybrids contain, in a background of mouse chromosomes, only the human X/autosome translocations which were present in the GM cells used for cell hybridization. Sublines of A9 cells were established containing the following autosomal segments: 1q23----1qter; 1q12----1pter; 3p12----3pter; 3q21----3qter; 11q13----11qter; 11q13----11pter; 11p11----11qter; 11q23----11pter; 12q24----12pter; 16q24----16pter; 17q11----17pter.


Asunto(s)
Cromosomas Humanos , Células Híbridas/citología , Translocación Genética , Cromosoma X , Animales , Fusión Celular , Línea Celular , Fibroblastos , Humanos , Hipoxantina Fosforribosiltransferasa/deficiencia , Cariotipificación , Ratones
17.
Microbiol Immunol ; 25(6): 565-74, 1981.
Artículo en Inglés | MEDLINE | ID: mdl-6168892

RESUMEN

Inoculation of Ehrlich ascites carcinoma cells (EAC) into the peritoneal cavities of outbred ddY mice induced interferon (IFN) in the circulation. The maximum titer (1,280 U) was obtained at 24 hr after inoculation. This induced IFN had the characteristics of type I IFN, i.e., stability at pH2 and lability at 56 C. An increase in natural killer cell (NK) activity was also observed for the first 3 days after inoculation. In addition, plasma lactate dehydrogenase (LDH) activity was elevated in these mice. Inoculation of ascitic fluid or serum of EAC-bearing mice into normal mice increased plasma LDH activity six- to sevenfold over normal levels and elevated activities persisted throughout the life of the mice. These results suggest that the LDH-elevating agent was responsible for IFN induction and for enhancing NK activity. Because lactate dehydrogenase-elevating virus (LDV) can be eliminated from tumor cells by passage in vitro, we attempted to grow EAC in tissue culture for several months and re-examined whether the inoculation of such cells could elevate plasma LDH activity induce IFN and enhance NK activity. The results showed that inoculation of the passaged cells had no effect on these activities in normal mice. Therefore, we concluded that the IFN inducer was LDV which contaminated the EAC and then enhanced the NK activity. N-tropic murine leukemia virus also contaminated EAC, but this virus was not responsible because cultured cells of EAC still shed this virus.


Asunto(s)
Carcinoma de Ehrlich/microbiología , Interferones/biosíntesis , Células Asesinas Naturales/inmunología , Virus Elevador de Lactato Deshidrogenasa/fisiología , Animales , Carcinoma de Ehrlich/patología , Células Cultivadas , L-Lactato Deshidrogenasa/sangre , Masculino , Ratones
18.
Tohoku J Exp Med ; 168(2): 167-8, 1992 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-1306300

RESUMEN

To identify the tumor suppressor gene on human chromosome 11p15, we generated mouse microcell hybrids containing small transferable chromosome 11p15 fragments, which we have termed "DNA superfragments". These hybrids will be used to identify which fragments contain a tumor suppressor gene by direct transfer of the fragments to tumor cells via microcell fusion.


Asunto(s)
Cromosomas Humanos Par 11/efectos de la radiación , Genes Supresores de Tumor , Animales , Clonación Molecular , Humanos , Ratones
19.
Symp Fundam Cancer Res ; 39: 45-56, 1986.
Artículo en Inglés | MEDLINE | ID: mdl-3321309

RESUMEN

We demonstrated previously that carcinogen-induced neoplastic transformation of Syrian hamster embryo (SHE) cells requires multiple steps. Normal, diploid SHE cells and carcinogen-induced preneoplastic cells were transfected with different oncogenes. The normal, early-passage cells were not transformed by the v-Ha-ras or v-myc oncogenes alone, but the two oncogenes combined caused tumors in nude mice and syngeneic hamsters. Cytogenetic analysis of the ras-plus-myc-induced tumors showed a nonrandom chromosome loss (monosomy of chromosome 15) in the ras/myc tumor cells. Tumorigenicity of the ras/myc tumor cells was suppressed following hybridization with normal SHE cells; reexpression of tumorigenicity at later passages correlated with loss of chromosome 15. The hybrid cells in which tumorigenicity was suppressed still expressed the ras and myc oncogenes. An early change in carcinogen-induced neoplastic progression of SHE cells is induction of immortality. At early passages, immortal cells retain the ability to suppress tumorigenicity in cell hybrids. This ability decreases with passaging of immortal cell lines. The susceptibility of immortal cell lines to neoplastic transformation by DNA transfection with the v-Ha-ras oncogene or tumor DNA inversely correlated with the tumor-suppressive ability of the cells in cell hybrids. These observations indicate that neoplastic transformation of SHE cells involves at least three steps: (1) induction of immortality, (2) activation of a transforming gene or oncogene, and (3) loss of or inactivation of a tumor-suppressor gene.


Asunto(s)
Neoplasias/genética , Oncogenes , Supresión Genética , Animales , Transformación Celular Neoplásica , Humanos , Modelos Genéticos
20.
Jpn J Cancer Res ; 80(5): 413-8, 1989 May.
Artículo en Inglés | MEDLINE | ID: mdl-2502516

RESUMEN

Normal human fibroblasts (MRC-5 or NTI-4) were transfected with pSV2-neo plasmid DNA. Fifty G418-resistant fibroblast clones were isolated and independently fused to mouse A9 cells. The cell hybrids were selected and isolated in the medium containing G418 plus ouabain. Since micronuclei were more efficiently induced in these hybrids compared to parental human fibroblasts by colcemid treatment, the transfer of neo-tagged human chromosomes in the hybrids to mouse A9 cells was performed via microcell fusion. Two hundred A9 microcell hybrids were isolated and karyotyped. Among them, thirteen microcell clones, each containing a single human chromosome 1, 2, 5, 6, 7, 8, 10, 11, 12, 15, 18, 19 or 20 were established. Isozyme analyses conformed the presence of each human chromosome in these A9 microcell clones. The results of Southern blot and chromosomal in situ hybridization analyses indicate that the human chromosomes in these clones were tagged with pSV2-neo plasmid DNA.


Asunto(s)
Cromosomas Humanos , Neomicina/farmacología , Factores R , Transfección , Animales , Fusión Celular , Humanos , Isoenzimas/análisis , Ratones
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda