Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Cytogenet Genome Res ; 125(1): 19-25, 2009.
Article in English | MEDLINE | ID: mdl-19617692

ABSTRACT

Xist non-coding RNA (ncRNA) is essential for X chromosome inactivation (XCI). Some genes can escape from XCI, but how this occurs is unknown. We developed a modified RNA tagging and recovery of associated proteins (TRAP) method to study the association between Xist RNA and its target genes. In mouse cells, Xist RNA was detected on the Uba1 gene, but not on Jarid1c and Utx genes, which escape from XCI. Using this technique we were able to show that the Xist RNA molecule is not present on active genes that escape from XCI, but is present on genes inactivated by XCI, suggesting that this method is a powerful tool for functional analysis of ncRNA.


Subject(s)
Chromatin/genetics , Chromatin/metabolism , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , X Chromosome Inactivation/genetics , X Chromosome/genetics , X Chromosome/metabolism , Animals , Cell Line , Chromatin Immunoprecipitation , DNA/genetics , DNA/metabolism , Female , Gene Expression , In Situ Hybridization , In Situ Hybridization, Fluorescence , Male , Mice , RNA, Long Noncoding , Reverse Transcriptase Polymerase Chain Reaction
2.
Mol Cell ; 7(4): 823-32, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11336705

ABSTRACT

The PKD1 gene accounts for 85% of autosomal dominant polycystic kidney disease (ADPKD), the most common human genetic disorder. Rats with a germline inactivation of one allele of the Tsc2 tumor suppressor gene developed early onset severe bilateral polycystic kidney disease, with similarities to the human contiguous gene syndrome caused by germline codeletion of PKD1 and TSC2 genes. Polycystic rat renal cells retained two normal Pkd1 alleles but were null for Tsc2 and exhibited loss of lateral membrane-localized polycystin-1. In tuberin-deficient cells, intracellular trafficking of polycystin-1 was disrupted, resulting in sequestration of polycystin-1 within the Golgi and reexpression of Tsc2 restored correct polycystin-1 membrane localization. These data identify tuberin as a determinant of polycystin-1 functional localization and, potentially, ADPKD severity.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Alleles , Animals , Cell Membrane/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Genes, Tumor Suppressor/physiology , Golgi Apparatus/metabolism , Proteins/genetics , Rats , TRPP Cation Channels , Transfection , Tuberous Sclerosis Complex 2 Protein , Tumor Cells, Cultured , Tumor Suppressor Proteins
3.
J Hum Genet ; 46(3): 137-45, 2001.
Article in English | MEDLINE | ID: mdl-11310581

ABSTRACT

As an in vitro assay system for the identification of human imprinted genes, a library of human/mouse A9 monochromosomal hybrids containing a single, intact bsr-tagged human chromosome of known parental origin, derived from normal human fibroblasts, has been previously generated by microcell-mediated chromosome transfer (MMCT). To supplement this assay system, we constructed additional 700 A9 monochromosomal hybrids, using a pSTneo or pPGKneo selection marker. To validate the A9 hybrids, we screened them with chromosome-specific polymorphic markers, and identified the hybrids containing either human chromosome 6, 7, 14, 18, or 21 of known parental origin. Matching paternal and maternal chromosome pairs of A9 hybrids were identified for chromosomes 6, 7, 14, and 18. The paternal-specific expression of ZAC (zinc finger protein, which regulates apoptosis and cell cycle arrest) and HYMAI (hydatidiform mole-associated and imprinted transcript), and the maternal-specific methylation of a CpG island within an imprinted domain on human chromosome 6q24, were maintained in A9 hybrids. For an example, we profiled the expression of expressed sequence tags (ESTs) and the methylation of CpG islands in the 300-kb imprinted domain around 6q24, which may be associated with cancers and transient neonatal diabetes mellitus (TNDM). Thus, the 700 A9 hybrids should be useful for various aspects of imprinting studies.


Subject(s)
Chromosomes, Human, Pair 6/genetics , Genomic Imprinting , Hybrid Cells , Animals , Base Sequence , CpG Islands , DNA Methylation , DNA Primers/genetics , Expressed Sequence Tags , Female , Genetic Techniques , Humans , In Situ Hybridization, Fluorescence , Infant, Newborn , Male , Mice
4.
Oncogene ; 20(7): 828-35, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11314017

ABSTRACT

Based on the sites of frequent allelic loss in hepatocellular carcinoma, five normal human chromosomes (2, 4, 5, 10 and 16) were transferred individually into a telomerase-positive human hepatocellular carcinoma cell line, Li7HM, by microcell-mediated chromosome transfer (MMCT). Chromosome 10, but not the others, repressed telomerase activity immediately and stopped cell growth after 50 population doublings (PDs). Loss of the transferred 10p loci resulted in the emergence of revertant cells that continued to proliferate and expressed telomerase activity, suggesting the presence of a telomerase repressor gene on this chromosomal arm. Transfer of a series of defined fragments from chromosome 10p successfully narrowed down the responsible region: a 28.9-cM region on 10p15 (between WI-4752 and D10S249), but not a 26.2-cM region (between D10S1728 and D10S249), caused repression of telomerase activity and progressive telomere shortening. A strong correlation between the expression level of telomerase catalytic subunit gene (hTERT) and telomerase activity was observed. These findings suggest that a novel telomerase repressor gene which controls the expression of hTERT is located on the 2.7-cM region (between WI-4752 and D10S1728) on chromosome 10p15.1.


Subject(s)
Chromosomes, Human, Pair 10/genetics , Repressor Proteins/genetics , Telomerase/genetics , Carcinoma, Hepatocellular , Chromosome Mapping , Humans , Hybrid Cells , In Situ Hybridization, Fluorescence , Liver Neoplasms , Tumor Cells, Cultured
5.
Biochem Biophys Res Commun ; 273(1): 219-24, 2000 Jun 24.
Article in English | MEDLINE | ID: mdl-10873590

ABSTRACT

Down syndrome (DS) leads to cardiac defects which are common and significant in babies with DS. We recently generated chimeric mice carrying a human chromosome (hChr) 21. The contribution ratio of embryonic stem (ES) cells containing a hChr 21 was specifically low in the heart, compared to other organs, and cardiovascular malformations were observed, suggesting that an additional copy of hChr 21 also disrupts the normal development of heart in mice. Here we describe that the presence of hChr 21 in ES cells delays the appearance of beating cardiomyocyte during differentiation, whereas differentiation into other cell types is not disrupted. Furthermore, the defect in cardiogenesis was restored following the deletion of a specific region of hChr 21. Therefore, we conclude that the imbalance of specific gene(s) on hChr 21 may lead to the disturbance of cardiogenesis and that this may be a useful system to model and investigate the cardiac defects of human DS.


Subject(s)
Chromosomes, Human, Pair 21/genetics , Heart Defects, Congenital/embryology , Heart Defects, Congenital/genetics , Myocardium/pathology , Stem Cells/pathology , Animals , Cartilage/cytology , Cartilage/metabolism , Cell Differentiation , Cell Line , Chromosome Deletion , Coculture Techniques , Gene Dosage , Heart Defects, Congenital/pathology , Heart Defects, Congenital/physiopathology , Humans , In Situ Hybridization, Fluorescence , Mice , Morphogenesis/genetics , Mutation/genetics , Myocardium/metabolism , Phenotype , Stem Cells/metabolism
6.
Cancer Genet Cytogenet ; 116(2): 158-63, 2000 Jan 15.
Article in English | MEDLINE | ID: mdl-10640149

ABSTRACT

Human chromosome 11 is expected to carry tumor suppressor genes for a variety of human cancers, including bladder carcinoma. To examine the functional role of a putative tumor suppressor gene(s) on this chromosome in the development of bladder carcinoma, we performed microcell-mediated transfer of chromosome 11 into the bladder carcinoma cell line, JTC-32. Fifteen of 20 colonies formed by the transfer experiment showed a remarkable change in cell morphology. They flattened and ceased growing, or senesced, prior to 10 population doublings. The presence of transferred chromosome 11-derived fragments in the growth-arrested cells was confirmed by PCR-based polymorphism analyses. The remaining 5 microcell hybrid clones exhibited a parental cell-like morphology, and presumably escaped from senescence, which was accompanied by deletions and/or rearrangements of the transferred chromosome 11. On the other hand, a transferred normal chromosome 7 neither changed the cell morphology nor arrested the cell growth. These results support the hypothesis that chromosome 11 contains a gene or genes which restore the senescence program lost during the immortalization process of JTC-32 cells.


Subject(s)
Cellular Senescence/genetics , Chromosomes, Human, Pair 11/genetics , Tumor Cells, Cultured/pathology , Urinary Bladder Neoplasms/pathology , Aged , Female , Humans , In Situ Hybridization, Fluorescence , Polymerase Chain Reaction , Urinary Bladder Neoplasms/genetics
7.
DNA Res ; 6(3): 165-72, 1999 Jun 30.
Article in English | MEDLINE | ID: mdl-10470847

ABSTRACT

To develop an systematic in vitro approach for the study of genomic imprinting, we generated a new library of human/mouse A9 monochromosomal hybrids. We used whole cell fusion and microcell-mediated chromosome transfer to generate A9 hybrids containing a single, intact, bsr-tagged human chromosome derived from primary fibroblasts. A9 hybrids were identified that contained either human chromosome 1, 2, 4, 5, 7, 8, 10, 11, 15, 18, 20, or X. The parental origin of these chromosomes was determined by polymorphic analysis using microsatellite markers, and matched hybrids containing maternal and paternal chromosomes were identified for chromosomes 5, 10, 11 and 15. The imprinted gene KVLQT1 on human chromosome 11p15.5 was expressed exclusively from the maternal chromosome in A9 hybrids, and the parental-origin-specific expression patterns of several other imprinted genes were also maintained. This library of human monochromosomal hybrids is a valuable resource for the mapping and cloning of human genes and is a novel in vitro system for the screening of imprinted genes and for their functional analysis.


Subject(s)
Chromosomes, Human/genetics , Gene Expression , Genomic Imprinting/genetics , Polymorphism, Genetic/genetics , Animals , Cell Line , Female , Humans , Hybrid Cells , In Situ Hybridization, Fluorescence , Karyotyping , Male , Mice , Microsatellite Repeats , Transfection
8.
Mol Carcinog ; 25(4): 249-55, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10449031

ABSTRACT

Maintenance of telomeres, commonly through expression of telomerase activity, is necessary but may not be sufficient for human cells to escape from the cellular senescence program and become immortal. We report here that human tumor cells could undergo cellular senescence in the presence of telomerase activity when a specific normal human chromosome was introduced via microcell-mediated chromosome transfer. The cell models studied include SiHa (uterine cervical carcinoma cells expressing E6 and E7 oncoproteins of human papillomavirus type 16) with a transferred chromosome 2, CC1 (choriocarcinoma cells expressing an amino-terminally truncated p53 protein) with a transferred chromosome 7, and JTC-32 (bladder carcinoma cells) with a transferred chromosome 11. The microcell hybrids with the indicated chromosomes ceased to divide after five to 10 population doublings and showed senescence-associated beta-galactosidase activity but still expressed the genes encoding three components of human telomerase, consistent with the retention of telomerase activity. These results are evidence for barriers to human cell immortalization, which involve activation of unidentified senescence-inducing genes that function independently of inactivation of telomerase.


Subject(s)
Cellular Senescence/genetics , Gene Transfer Techniques , Telomerase/metabolism , Cell Line, Transformed , Humans , Hybrid Cells , Telomerase/genetics
9.
Hum Mol Genet ; 8(7): 1209-17, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10369866

ABSTRACT

Mammalian imprinted genes are frequently arranged in clusters on particular chromosomes. The imprinting cluster on human chromosome 11p15 is associated with Beckwith-Wiedemann syndrome (BWS) and a variety of human cancers. To clarify the genomic organization of the imprinted cluster, an extensive screen for differentially expressed transcripts in the 11p15 region was performed using monochromosomal hybrids with a paternal or maternal human chromosome 11. Here we describe an imprinted antisense transcript identified within the KvLQT1 locus, which is associated with multiple balanced chromosomal rearrangements in BWS and an additional breakpoint in embryonal rhabdoid tumors. The transcript, called LIT1 (long QT intronic transcript 1), was expressed preferentially from the paternal allele and produced in most human tissues. Methylation analysis revealed that an intronic CpG island was specifically methylated on the silent maternal allele and that four of 13 BWS patients showed complete loss of maternal methylation at the CpG island, suggesting that antisense regulation is involved in the development of human disease. In addition, we found that eight of eight Wilms' tumors exhibited normal imprinting of LIT1 and five of five tumors displayed normal differential methylation at the intronic CpG island. This contrasts with five of six tumors showing loss of imprinting of IGF2. We conclude that the imprinted gene domain at the KvLQT1 locus is discordantly regulated in cancer from the imprinted domain at the IGF2 locus. Thus, this positional approach using human monochromosomal hybrids could contribute to the efficient identification of imprinted loci in humans.


Subject(s)
Beckwith-Wiedemann Syndrome/genetics , Membrane Proteins , Potassium Channels, Voltage-Gated , Potassium Channels/genetics , RNA, Antisense/genetics , Animals , Base Sequence , CpG Islands/genetics , DNA Methylation , Fibroblasts/physiology , Genomic Imprinting , Humans , Hybrid Cells , Introns , KCNQ Potassium Channels , KCNQ1 Potassium Channel , Mice , Molecular Sequence Data , Potassium Channels/biosynthesis , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Sequence Alignment , Wilms Tumor/genetics
10.
Genes Chromosomes Cancer ; 24(1): 1-8, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9892102

ABSTRACT

Allelotype analyses of human prostate cancer indicate that allelic losses on human chromosome arms 7q, 8p, 10q, 13q, 16q, 17q, and 18q are observed frequently. For the study of the possible biological significance of the frequently observed deletions on chromosome arm 7q in human prostate cancer, human chromosome 7 was introduced into highly metastatic rat prostate cancer cells by use of a microcell-mediated chromosome transfer technique. The introduction of human chromosome 7 resulted in the suppression of metastatic ability of the microcell hybrids, whereas no suppression of tumorigenicity was observed. To identify the portion of chromosome 7 containing the metastasis-suppressive function gene, the derivative chromosome 7 that was generated with the initial transfer was retransferred into rat prostate cancer cells. Human chromosome 7-containing rat prostate cancer cells could be used as the donor cells, because rodent cells produced a sufficient number of microcells with colchicine treatment. Cytogenetic and molecular analyses of these clones demonstrated that loss of segments on 7q was related to the reexpression of the metastatic phenotype. These results show that human 7q contains a metastasis suppressor gene or genes for rat prostate cancer. The findings also suggest that this gene may play an important role in the progression of human prostate cancer.


Subject(s)
Chromosomes, Human, Pair 7/genetics , Genes, Tumor Suppressor/genetics , Lung Neoplasms/secondary , Prostatic Neoplasms/genetics , Animals , Chromosome Mapping , Genetic Markers , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Lung Neoplasms/chemistry , Lung Neoplasms/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Prostatic Neoplasms/chemistry , Rats , Tumor Cells, Cultured
11.
Genes Chromosomes Cancer ; 23(2): 123-33, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9739015

ABSTRACT

Telomeres, which are the repeated sequences located on both ends of chromosomes in eukaryotes, are known to shorten with each cell division, and their eventual loss is thought to result in cellular senescence. Unlike normal somatic cells, most tumor cells show activation of telomerase, a ribonucleoprotein enzyme that stably maintains telomere length by addition of the sequences of TTAGGG repeats to telomeres. The KC12 cell line derived from a renal cell carcinoma in a patient with von Hippel-Lindau disease showed telomerase activity and loss of heterozygosity on the short arm of chromosome 3. Introduction of a normal human chromosome 3 into KC12 cells by microcell fusion induced cellular senescence, accompanied by suppression of telomerase activity and shortening of telomere length. Microcell hybrids that escaped from cellular senescence maintained telomere length and telomerase activity similar to those of the parental KC12 cells. We previously showed a similar suppression of telomerase activity by introduction of chromosome 3 into another renal cell carcinoma cell line, RCC23. The putative telomerase repressor gene was mapped to chromosome region 3p14.2-p21.1 by deletion mapping of KC12 + chromosome 3 revertants that escaped from cellular senescence and by transfer of subchromosomal fragments of chromosome 3 into RCC23 cells.


Subject(s)
Chromosomes, Human, Pair 3/genetics , Repressor Proteins/genetics , Telomerase/genetics , Carcinoma, Renal Cell/enzymology , Carcinoma, Renal Cell/pathology , Clone Cells/enzymology , Clone Cells/pathology , Humans , Hybrid Cells/enzymology , Hybrid Cells/pathology , Kidney Neoplasms/enzymology , Kidney Neoplasms/pathology , Restriction Mapping , Telomerase/metabolism , Tumor Cells, Cultured/enzymology , Tumor Cells, Cultured/pathology
12.
Genes Cells ; 3(4): 245-55, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9663659

ABSTRACT

BACKGROUND: Genomic imprinting in mammals is thought to result from epigenetic modifications to chromosomes during gametogenesis, which leads to differential allelic expression during development. There is a requirement for an appropriate experimental system to enable the analysis of the mechanisms of genomic imprinting during embryogenesis. RESULTS: To develop a novel in vitro system for studying the molecular basis of genomic imprinting, we constructed mouse cell lines containing either a paternal or maternal human chromosome 11, by microcell-mediated chromosome transfer. Allele-specific expression and DNA methylation studies revealed that the imprinting status of the human H19 gene was maintained in mouse A9 mono-chromosomal hybrids. Each parental human chromosome was introduced independently into mouse near-diploid immortal fibroblasts (m5S) and two embryonal carcinoma (EC) cell lines (OTF9-63 and P19). The paternal allele of human H19 remained in a repressed state in m5S cells, but was de-repressed in both EC cells. The paternal H19 allele was demethylated extensively in OTF9-63 cells, whereas the only alteration in P19 hybrids was de novo methylation on both alleles in the 3' region. Following in vitro differentiation, the expressed paternal H19 allele was selectively repressed in differentiated derivatives of EC hybrids. CONCLUSION: These results indicated that human imprint marks could function effectively in mouse cells, and that the imprinting process was epigenetically reprogrammed in embryonal carcinoma cells, without erasure of the primary imprint that marked the parental origin. Therefore, these mono-chromosomal hybrids could provide a valuable in vitro system to study the mechanisms involved in the regulation of imprinted gene expression.


Subject(s)
Gene Expression Regulation , Genomic Imprinting , Muscle Proteins/genetics , RNA, Untranslated , Animals , Cell Differentiation , Cell Fusion , Chromosomes, Human, Pair 11/genetics , DNA Methylation , Female , Humans , Hybrid Cells , Male , Mice , RNA, Long Noncoding , Species Specificity
13.
Nihon Rinsho ; 56(5): 1108-14, 1998 May.
Article in Japanese | MEDLINE | ID: mdl-9613105

ABSTRACT

Genes involved in the senescence program have been mapped to over ten different genetic loci using microcell fusion to introduced human chromosomes and restore the senescence program. Multiple pathways of cellular senescence have also been demonstrated by chromosome transfer, indicating that the functions of the mapped senescence genes are probably difference. Restoration of cellular senescence by chromosome 3p or 10p is associated with repression of telomerase function in human tumor cell lines, and other chromosomes induced cellular senescence without telomerase suppression. This evidence suggests that telomerase suppression and thereby telomere shortening is one of several pathways involved in cellular senescence.


Subject(s)
Cellular Senescence/physiology , Telomerase/genetics , Animals , Humans , Repressor Proteins/physiology , Telomerase/physiology
14.
Mol Carcinog ; 22(1): 34-45, 1998 May.
Article in English | MEDLINE | ID: mdl-9609099

ABSTRACT

To identify the subchromosomal region that carries the cellular-senescence-restoring program of the human cervical carcinoma cell line SiHa, we constructed by irradiation microcell-mediated chromosome transfer a library of mouse A9 cells containing various fragments of human chromosome 2 tagged with pSV2neo in 2p11-p12. Eighty-seven clones were isolated and screened for the presence of human sequences by inter-Alu and inter-L1 polymerase chain reaction (PCR), and six clones exhibiting PCR-laddering patterns that differed from those of the A9 cells containing an intact chromosome 2 were examined further. Chromosome analysis and fluorescence in situ hybridization (FISH) using human-specific repetitive sequences revealed that four of these clones contained single subchromosomal transferable fragments (STFs). Southern blot hybridization of 14 cosmid markers revealed that the STFs in A9 cells were derived from human chromosome 2. These STFs were transferred into SiHa cells by microcell fusion, and one of the STFs restored the cellular-senescence program. The concordance of the cellular-senescence-restoring program with the presence or absence of specific DNA fragments of chromosome 2 indicated that the putative cellular-senescence gene was located in 2q32-qter. For more detailed mapping, we constructed mouse A9 cells containing STFs derived from human chromosome 2 tagged with pSTneo at different regions in 2q31-qter. PCR-laddering and FISH analyses were used to identify six clones that contained different STFs. These STFs were transferred into SiHa cells, and one of the three clones that restored cellular senescence contained a small fragment of human chromosome 2. This STF was shown by PCR analysis using 14 human chromosome 2-specific primer pairs to be smaller than 12.2 cM and was mapped to the 2q37 region by FISH analysis with inter-Alu PCR. Beta-galactosidase activity, which is a biomarker of senescent cells, and telomerase activity similar to that found in parental SiHa cells were detected in SiHa microcell hybrids, suggesting that the putative cellular-senescence gene was not involved in a telomerase pathway but rather in an alternate pathway of cellular senescence.


Subject(s)
Cellular Senescence/genetics , Chromosome Mapping/methods , Chromosomes, Human, Pair 2 , Gene Transfer Techniques , Animals , Cell Line , Cosmids , DNA Primers , Female , Fluorescent Dyes , Genetic Markers , Humans , In Situ Hybridization, Fluorescence/methods , Karyotyping , Mice , Polymerase Chain Reaction/methods , Sequence Tagged Sites , Tumor Cells, Cultured , Uterine Cervical Neoplasms
15.
Mol Carcinog ; 21(1): 17-25, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9473768

ABSTRACT

Human telomerase, a ribonucleoprotein that adds TTAGGG repeats onto telomeres and compensates for their shortening, is repressed in most normal human somatic cells. Human somatic cells are considered to have a limited proliferation capacity because of the telomere shortening. Although immortalization of somatic cells is often associated with telomerase reactivation, there are some immortal cells in which telomerase activity is undetectable. In these cells, telomeres may be maintained by an unknown mechanism other than telomerase reactivation. To examine the genetic regulation of telomerase activity, we constructed hybrids between immortal cells with (HepG2) and without (KMST6) telomerase activity. These two cell lines had relatively short and long telomeres, respectively. The hybrid cells continued to proliferate without detectable telomerase activity even after 100 population doublings. Telomerase-positive subpopulations occasionally appeared after serial passages. Southern blot analysis revealed that the hybrids had long terminal restriction fragments similar to that of KMST6, regardless of telomerase activity, and fluorescence in situ hybridization with a telomeric probe showed high-intensity hybridization signals on telomeres, indicating relatively long telomeric repeats. These results suggest that the telomerase-negative immortal cells contain a gene or genes functioning as a telomerase repressor and maintain telomere length by a dominant mechanism other than telomerase reactivation.


Subject(s)
Telomerase/biosynthesis , Telomerase/genetics , Telomere/genetics , Cells, Cultured , Enzyme Repression , Female , Fibroblasts/enzymology , Humans , Hybrid Cells/cytology , Hybrid Cells/enzymology , Karyotyping , Telomerase/metabolism
16.
Hum Mol Genet ; 6(12): 2127-33, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9328477

ABSTRACT

We have constructed mouse A9 hybrids containing a single normal human chromosome 15, via microcell-mediated chromosome transfer. Cytogenetic and DNA-polymorphic analyses identified mouse A9 hybrids that contained either a paternal or maternal human chromosome 15. Paternal specific expression of the known imprinted genes SNRPN (small nuclear ribonucleoprotein-associated polypeptide N gene) and IPW (imprinted gene in the Prader-Willi syndrome region) was maintained in the A9 hybrids. Using this system, we first demonstrated that human GABAAreceptor subunit genes, GABRB3 , GABRA5 and GABRG3 , were expressed exclusively from the paternal allele and that E6-AP (E6-associated protein or UBE3A ) was biallelically expressed. Moreover, the 5' portion of the GABRB3 gene was found to be hypermethylated on the paternal allele. Our data imply that GABAAreceptor subunit genes are imprinted and are possible candidates for Prader-Willi syndrome, and that this human monochromosomal hybrid system enables the efficient analysis of imprinted loci.


Subject(s)
Chromosomes, Human, Pair 15/genetics , Genomic Imprinting , Receptors, GABA-A/genetics , Adult , Animals , Autoantigens/chemistry , Autoantigens/genetics , Cell Separation , Cells, Cultured , Female , Fibroblasts , Gene Expression Regulation , Gene Transfer Techniques , Humans , Hybrid Cells , Ligases/genetics , Male , Mice , Polymerase Chain Reaction , Prader-Willi Syndrome/genetics , Ribonucleoproteins, Small Nuclear/genetics , Ubiquitin-Protein Ligases , snRNP Core Proteins
17.
Nat Genet ; 16(2): 133-43, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9171824

ABSTRACT

Human chromosomes or chromosome fragments derived from normal fibroblasts were introduced into mouse embryonic stem (ES) cells via microcell-mediated chromosome transfer (MMCT) and viable chimaeric mice were produced from them. Transferred chromosomes were stably retained, and human genes, including immunoglobulin (Ig) kappa, heavy, lambda genes, were expressed in proper tissue-specific manner in adult chimaeric tissues. In the case of a human chromosome (hChr.) 2-derived fragment, it was found to be transmitted to the offspring through the germline. Our study demonstrates that MMCT allows for introduction of very large amounts of foreign genetic material into mice. This novel procedure will facilitate the functional analyses of human genomes in vivo.


Subject(s)
Chimera , Chromosomes, Human , Gene Transfer Techniques , Germ-Line Mutation , Animals , Cell Fusion , Female , Genome, Human , Humans , Immunoglobulins/genetics , Male , Mice , RNA, Messenger/genetics , Stem Cells
18.
Hum Mol Genet ; 6(13): 2243-6, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9361029

ABSTRACT

The Wilms' tumor suppressor gene ( WT1 ) was previously identified as being imprinted, with frequent maternal expression in human placentae and fetal brains. We examined the allele-specific expression of WT1 in cultured human fibroblasts from 15 individuals. Seven of 15 fibroblast lines were heterozygous for polymorphic alleles, and the expression patterns were variable, i.e., equal, unequal or monoallelic paternal expression in three, two and two cases, respectively. Exclusive paternal expression of WT1 was also shown in non-cultured peripheral lymphocytes from the latter two individuals. The allele-specific expression profiles of other imprinted genes, IGF2 and H19, on human chromosome 11 were constant and consistent with those in other tissues. Our unexpected observations of paternal or biallelic expression of WT1 in fibroblasts and lymphocytes, together with the previous findings of maternal or biallelic expression in placentae and brains, suggest that the allele-specific regulatory system of WT1 is unique and may be controlled by a putative tissue- and individual-specific modifier.


Subject(s)
DNA-Binding Proteins/biosynthesis , Fibroblasts/metabolism , Gene Expression Regulation , Genes, Wilms Tumor , Genomic Imprinting , Lymphocytes/metabolism , RNA, Untranslated , Transcription Factors/biosynthesis , Adult , Aged , Alleles , Cells, Cultured , Chromosomes, Human, Pair 11/genetics , Dinucleotide Repeats , Female , Fetus/metabolism , Humans , Infant, Newborn , Insulin-Like Growth Factor II/biosynthesis , Insulin-Like Growth Factor II/genetics , Male , Middle Aged , Muscle Proteins/biosynthesis , Muscle Proteins/genetics , Organ Specificity , Polymerase Chain Reaction , RNA, Long Noncoding , WT1 Proteins
19.
Genes Chromosomes Cancer ; 17(4): 260-8, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8946208

ABSTRACT

Our previous studies demonstrated that human chromosome 8 contains metastasis suppressor gene(s) for rat prostate cancer. However, it is still unknown which portion of human chromosome 8 is associated with suppression of metastatic ability, because all of the clones in which metastatic ability is suppressed contain at least one copy of intact human chromosome 8. In the present study, we used the irradiated microcell-mediated chromosome transfer technique to enrich for specific chromosomal arm deletions of selected chromosomes. The resultant series of human chromosomes 8 with a variety of chromosomal deletions was introduced into highly metastatic Dunning rat prostate cancer cells. All of the resultant microcell hybrids showed reduced metastatic ability. To obtain a smaller size of human chromosome 8 and to locate further the region of metastasis suppressor gene(s), the most reduced size of human chromosome 8 that was generated with the initial irradiated chromosome transfer was retransferred into the Dunning cancer cells without irradiation. The resultant microcell hybrids were analyzed to determine which portion of human chromosome 8 suppressed the metastatic ability of the recipient cells. This analysis demonstrates that the portion of human chromosome 8 containing metastasis suppressor gene(s) for rat prostate cancer cells lies on human chromosome segment 8p21-p12, where frequent allelic losses have been detected in allelotype analyses of human prostate cancer. This suggests that one of the metastasis suppressor genes for rat prostate cancer on human chromosome 8 may also play an important role in the progression of human prostate cancer.


Subject(s)
Chromosome Mapping , Chromosomes, Human, Pair 8 , Gene Transfer Techniques , Genes, Tumor Suppressor/genetics , Prostatic Neoplasms/genetics , Animals , Chromosome Banding , Humans , In Situ Hybridization, Fluorescence , Male , Polymerase Chain Reaction , Rats , Tumor Cells, Cultured
20.
Hum Cell ; 9(4): 301-8, 1996 Dec.
Article in English | MEDLINE | ID: mdl-9183662

ABSTRACT

Hybrids between immortal cells and normal cells senesce, indicating that immortal cells have lost, mutated or inactivated genes that are required for the program of senescence in normal cells. Genes involved in the senescence program have been mapped to over 10 different genetic loci by introduction of normal human chromosomes via microcell fusion. Multiple pathways of cellular senescence have also been demonstrated by chromosome transfer, indicating that the functions of the mapped senescence genes are probably different. One possibility is that one or more of these senescence genes may suppress telomerase activity in immortal cells, resulting in telomere shortening and cellular senescence. To test this hypothesis, telomerase activity and the length of terminal restriction fragments (TRFs) have been examined in microcell hybrids. The loss of indefinite growth potential was either with or without the loss of telomerase activity activity and shortening of telomeres in the microcell hybrids containing the introduced chromosome. The findings suggest that telomerase regulation is one of multiple pathways to cellular senescence.


Subject(s)
Cellular Senescence/genetics , Gene Expression Regulation, Enzymologic/physiology , Telomerase/genetics , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...