Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Clin Genet ; 80(3): 243-55, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21261604

ABSTRACT

Childhood brain tumours may be due to germline bi-allelic mismatch repair (MMR) gene mutations in MLH1, MSH2, MSH6 or PMS2. These mutations can also lead to colorectal neoplasia and haematological malignancies. Here, we review this syndrome and present siblings with early-onset rectal adenoma and papillary glioneural brain tumour, respectively, due to novel germline bi-allelic PMS2 mutations. Identification of MMR protein defects can lead to early diagnosis of this condition. In addition, assays for these defects may help to classify brain tumours for research protocols aimed at targeted therapies.


Subject(s)
Adenoma/genetics , Adenosine Triphosphatases , Brain Neoplasms/genetics , Colorectal Neoplasms/genetics , DNA Repair Enzymes , DNA-Binding Proteins , Germ-Line Mutation , Glioma/genetics , Adenoma/diagnosis , Adenoma/pathology , Adenosine Triphosphatases/genetics , Age of Onset , Brain Neoplasms/diagnosis , Brain Neoplasms/pathology , Child , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/pathology , DNA Mismatch Repair , DNA Mutational Analysis , DNA Repair Enzymes/genetics , DNA-Binding Proteins/genetics , Female , Glioma/diagnosis , Glioma/pathology , Heterozygote , Humans , Male , Microsatellite Repeats , Mismatch Repair Endonuclease PMS2 , Pedigree , Siblings , Syndrome , Young Adult
2.
Gene Ther ; 18(3): 213-9, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21160530

ABSTRACT

Gene targeting by single-stranded oligodeoxyribonucleotides (ssODNs) is a promising technique for introducing site-specific sequence alterations without affecting the genomic organization of the target locus. Here, we discuss the significant progress that has been made over the last 5 years in unraveling the mechanisms and reaction parameters underlying ssODN-mediated gene targeting. We will specifically focus on ssODN-mediated gene targeting in murine embryonic stem cells (ESCs) and the impact of the DNA mismatch repair (MMR) system on the targeting process. Implications of novel findings for routine application of ssODN-mediated gene targeting and challenges that need to be overcome for future therapeutic applications are highlighted.


Subject(s)
DNA Mismatch Repair/genetics , Embryonic Stem Cells , Gene Targeting/methods , Oligodeoxyribonucleotides/therapeutic use , Animals , Mice , Oligodeoxyribonucleotides/genetics
3.
Oncogene ; 27(8): 1142-54, 2008 Feb 14.
Article in English | MEDLINE | ID: mdl-17700522

ABSTRACT

In the absence of mitogenic stimuli, cells normally arrest in G(1/0), because they fail to pass the G1-restriction point. However, abrogation of the G1-restriction point (by loss of the retinoblastoma gene family) reveals a second-restriction point that arrests cells in G2. Serum-starvation-induced G2 arrest is effectuated through inhibitory interactions of p27(KIP1) and p21(CIP1) with cyclins A and B1 and can be reversed through mitogen re-addition. In this study, we have investigated the pathways that allow cell cycle re-entry from this G2 arrest. We provide evidence that recovery from G2 arrest depends on the rat sarcoma viral oncogene (RAS) and phosphatidylinositol-3 kinase pathways and show that oncogenic hits, such as overexpression of c-MYC or mutational activation of RAS can abrogate the G2-restriction point. Together, our results provide new mechanistic insight into multistep carcinogenesis.


Subject(s)
G2 Phase/physiology , Oncogenes/physiology , Signal Transduction/physiology , Animals , Cell Line , Cell Transformation, Neoplastic/genetics , Gene Expression Profiling , Genes, ras/physiology , Mice , Multigene Family/physiology , Phosphatidylinositol 3-Kinases/physiology , ras Proteins/physiology
4.
Gene Ther ; 13(8): 686-94, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16437133

ABSTRACT

We have previously demonstrated that site-specific insertion, deletion or substitution of one or two nucleotides in mouse embryonic stem cells (ES cells) by single-stranded deoxyribo-oligonucleotides is several hundred-fold suppressed by DNA mismatch repair (MMR) activity. Here, we have investigated whether compound mismatches and larger insertions escape detection by the MMR machinery and can be effectively introduced in MMR-proficient cells. We identified several compound mismatches that escaped detection by the MMR machinery to some extent, but could not define general rules predicting the efficacy of complex base-pair substitutions. In contrast, we found that four-nucleotide insertions were largely subject to suppression by the MSH2/MSH3 branch of MMR and could be effectively introduced in Msh3-deficient cells. As these cells have no overt mutator phenotype and Msh3-deficient mice do not develop cancer, Msh3-deficient ES cells can be used for oligonucleotide-mediated gene disruption. As an example, we present disruption of the Fanconi anemia gene Fancf.


Subject(s)
Fanconi Anemia/therapy , Genetic Therapy/methods , Mutagenesis, Insertional/methods , Oligonucleotides/genetics , Proteins/genetics , Stem Cells/metabolism , Animals , Base Pair Mismatch , Base Sequence , Cells, Cultured , Dimerization , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Mice , Mice, Knockout , Molecular Sequence Data , MutS Homolog 2 Protein/genetics , MutS Homolog 2 Protein/metabolism , MutS Homolog 3 Protein , Proteins/metabolism
5.
J Hered ; 93(3): 201-5, 2002.
Article in English | MEDLINE | ID: mdl-12195036

ABSTRACT

Meiotic recombination was studied in DNA mismatch repair (MMR)-deficient mice using a strain carrying a Pms2 knockout mutation. Using single-sperm typing, recombination was analyzed over five intervals on four chromosomes in four Pms2 -/- animals. A total of 1936 meioses were studied and compared to 1848 meioses from three Pms2 +/+ controls. A smaller study was carried out on a single interval in each of two chromosomes in an MMR-deficient mouse homozygous for the Msh2 knockout mutation. A total of 792 meioses were examined in the Msh2 -/- and 880 meioses in the Msh2 +/+ animal. Recombination fractions were not significantly different in either of the MMR-deficient mouse strains when compared to MMR-proficient controls. Our results appear to conflict with mouse embryonic stem (ES) cell gene-targeting experiments where MMR plays a major role in determining the efficiency of homologous recombination between nonidentical sequences. A number of possibilities could explain the apparent lack of a significant effect on meiosis.


Subject(s)
Base Pair Mismatch , DNA Repair Enzymes , Meiosis/genetics , Mice, Knockout/genetics , Adenosine Triphosphatases/deficiency , Adenosine Triphosphatases/genetics , Animals , Base Sequence , DNA Primers , DNA Repair/genetics , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Male , Mice , Mismatch Repair Endonuclease PMS2 , MutS Homolog 2 Protein , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Recombination, Genetic , Spermatozoa/abnormalities
6.
Mol Cell Biol ; 21(17): 5806-14, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11486020

ABSTRACT

The "pocket" proteins pRb, p107, and p130 are a family of negative growth regulators. Previous studies have demonstrated that overexpression of pRb can repress transcription by RNA polymerase (Pol) I. To assess whether pRb performs this role under physiological conditions, we have examined pre-rRNA levels in cells from mice lacking either pRb alone or combinations of the three pocket proteins. Pol I transcription was unaffected in pRb-knockout fibroblasts, but specific disruption of the entire pRb family deregulated rRNA synthesis. Further analysis showed that p130 shares with pRb the ability to repress Pol I transcription, whereas p107 is ineffective in this system. Production of rRNA is abnormally elevated in Rb(-/-) p130(-/-) fibroblasts. Furthermore, overexpression of p130 can inhibit an rRNA promoter both in vitro and in vivo. This reflects an ability of p130 to bind and inactivate the upstream binding factor, UBF. The data imply that rRNA synthesis in living cells is subject to redundant control by endogenous pRb and p130.


Subject(s)
Phosphoproteins/metabolism , Pol1 Transcription Initiation Complex Proteins , Proteins , RNA Polymerase I/metabolism , RNA, Ribosomal/biosynthesis , Retinoblastoma Protein/metabolism , 3T3 Cells , Animals , Cells, Cultured , DNA-Binding Proteins/metabolism , Mice , Nuclear Proteins/metabolism , Phosphoproteins/genetics , Retinoblastoma Protein/genetics , Retinoblastoma-Like Protein p107 , Retinoblastoma-Like Protein p130 , Transcription Factors/metabolism , Transcription, Genetic
8.
Genes Dev ; 14(23): 3051-64, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-11114893

ABSTRACT

The retinoblastoma suppressor pRB belongs to the family of so-called pocket proteins, which also includes p107 and p130. These proteins may functionally overlap in cell cycle control and tumor suppression. We have generated an isogenic set of embryonic stem (ES) cell lines carrying single or compound loss-of-function mutations in the Rb gene family, including a cell line completely devoid of all three pocket proteins. None of the knockout combinations affected the growth characteristics of ES cells; however, concomitant ablation of all three pocket proteins strongly impaired their differentiation capacity. For the generated genotypes, primary mouse embryonic fibroblasts (MEFs) also were obtained. While inactivation of Rb alone did not alleviate the senescence response of MEFs, pRB/p107-deficient MEFs, after having adapted to in vitro culturing, continued to proliferate at modest rate. Additional ablation of p130 rendered MEFs completely insensitive to senescence-inducing signals and strongly increased their proliferation rate. Although triple-knockout MEFs retained anchorage dependence, they lacked proper G(1) control and showed increased cell turnover under growth-inhibiting conditions.


Subject(s)
Cell Differentiation/physiology , Nuclear Proteins/physiology , Phosphoproteins/physiology , Retinoblastoma Protein/physiology , Animals , Cell Adhesion , Cell Division , Cell Line , G1 Phase , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Proteins/metabolism , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Retinoblastoma-Like Protein p107 , Retinoblastoma-Like Protein p130 , Stem Cells/cytology , Teratocarcinoma , Tumor Suppressor Protein p14ARF , Tumor Suppressor Protein p53/metabolism
9.
Oncogene ; 19(42): 4822-7, 2000 Oct 05.
Article in English | MEDLINE | ID: mdl-11039898

ABSTRACT

MYC transcription factors are potent stimulators of cell proliferation. It has been suggested that the CDK-inhibitor p27kip1 is a critical G1 phase cell cycle target of c-MYC. We show here that mouse embryo fibroblasts deficient for both p27kip1 and the related p21cip1 are still responsive to stimulation by c-MYC and can be arrested in G1 by a dominant negative mutant of c-MYC. This growth arrest can be overruled by ectopic expression of E2F or adenovirus E1A, but not by a mutant of E1A defective for binding to retinoblastoma family proteins. We show that fibroblasts with a genetic disruption of all three retinoblastoma family members (pRb, p107 and p130) are unresponsive to a dominant negative c-MYC mutant. These data indicate that p27kip1 is not the only rate limiting cell cycle target of c-MYC and suggest that regulation of E2F is also essential for c-MYC's mitogenic activity.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , Cell Cycle/physiology , DNA-Binding Proteins , Genes, cdc , Microtubule-Associated Proteins/physiology , Proteins , Proto-Oncogene Proteins c-myc/physiology , Tumor Suppressor Proteins , Adenovirus E1A Proteins/genetics , Adenovirus E1A Proteins/physiology , Animals , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclins/physiology , E2F Transcription Factors , Fibroblasts , G1 Phase/genetics , G1 Phase/physiology , Genes, Dominant , Genes, Retinoblastoma , Genes, myc , Mice , Mice, Knockout , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Phosphoproteins/deficiency , Phosphoproteins/genetics , Phosphoproteins/physiology , Recombinant Fusion Proteins/physiology , Retinoblastoma Protein/deficiency , Retinoblastoma Protein/genetics , Retinoblastoma Protein/physiology , Retinoblastoma-Binding Protein 1 , Retinoblastoma-Like Protein p107 , Retinoblastoma-Like Protein p130 , S Phase/genetics , S Phase/physiology , Transcription Factor DP1 , Transcription Factors/genetics , Transcription Factors/physiology
10.
Toxicol Lett ; 112-113: 245-50, 2000 Mar 15.
Article in English | MEDLINE | ID: mdl-10720738

ABSTRACT

Hemizygosity for genes that are essential for DNA mismatch repair (MMR) was found to underlie cancer predisposition in hereditary nonpolypsis colorectal cancer (HNPCC). Loss of the wild-type allele generates a MMR-deficient cell compartment with a high propensity to oncogenic transformation. MMR deficiency not only accelerates spontaneous mutagenesis resulting from DNA replication errors, but also affects the cellular response to genotoxic agents. To study the consequences of MMR deficiency in vitro and to provide experimental access to HNPCC we have generated MMR-deficient cell lines and mice. The combination of MMR deficiency and exposure to genotoxic agents strongly accelerated lymphomagenesis.


Subject(s)
Base Pair Mismatch/genetics , DNA-Binding Proteins , Lymphoma/genetics , Mutagens/toxicity , Proto-Oncogene Proteins/genetics , Animals , Cells, Cultured , DNA Repair/genetics , Genetic Predisposition to Disease , Lymphoma/chemically induced , Mice , MutS Homolog 2 Protein
11.
Nat Genet ; 23(3): 359-62, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10545954

ABSTRACT

Cancer predisposition in hereditary non-polyposis colon cancer (HNPCC) is caused by defects in DNA mismatch repair (MMR). Mismatch recognition is attributed to two heterodimeric protein complexes: MutSalpha (refs 2, 3, 4, 5), a dimer of MutS homologues MSH2 and MSH6; and MutSbeta (refs 2,7), a dimer of MSH2 and MSH3. These complexes have specific and redundant mismatch recognition capacity. Whereas MSH2 deficiency ablates the activity of both dimers, causing strong cancer predisposition in mice and men, loss of MSH3 or MSH6 (also known as GTBP) function causes a partial MMR defect. This may explain the rarity of MSH6 and absence of MSH3 germline mutations in HNPCC families. To test this, we have inactivated the mouse genes Msh3 (formerly Rep3 ) and Msh6 (formerly Gtmbp). Msh6-deficient mice were prone to cancer; most animals developed lymphomas or epithelial tumours originating from the skin and uterus but only rarely from the intestine. Msh3 deficiency did not cause cancer predisposition, but in an Msh6 -deficient background, loss of Msh3 accelerated intestinal tumorigenesis. Lymphomagenesis was not affected. Furthermore, mismatch-directed anti-recombination and sensitivity to methylating agents required Msh2 and Msh6, but not Msh3. Thus, loss of MMR functions specific to Msh2/Msh6 is sufficient for lymphoma development in mice, whereas predisposition to intestinal cancer requires loss of function of both Msh2/Msh6 and Msh2/Msh3.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , DNA Repair/genetics , DNA-Binding Proteins/genetics , Gene Deletion , Genetic Predisposition to Disease/genetics , Multidrug Resistance-Associated Proteins , Alleles , Animals , Base Pair Mismatch/genetics , Cell Death/drug effects , Colorectal Neoplasms, Hereditary Nonpolyposis/metabolism , Colorectal Neoplasms, Hereditary Nonpolyposis/mortality , Colorectal Neoplasms, Hereditary Nonpolyposis/pathology , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Female , Incidence , Male , Methylnitronitrosoguanidine/toxicity , Mice , Mice, Transgenic , MutS Homolog 3 Protein , Mutagenesis, Insertional , Oligodeoxyribonucleotides/genetics , Oligodeoxyribonucleotides/metabolism , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism , Survival Rate , Time Factors
12.
Genes Dev ; 13(5): 523-31, 1999 Mar 01.
Article in English | MEDLINE | ID: mdl-10072381

ABSTRACT

Members of the mammalian mismatch repair protein family of MutS and MutL homologs have been implicated in postreplicative mismatch correction and chromosome interactions during meiotic recombination. Here we demonstrate that mice carrying a disruption in MutS homolog Msh5 show a meiotic defect, leading to male and female sterility. Histological and cytological examination of prophase I stages in both sexes revealed an extended zygotene stage, characterized by impaired and aberrant chromosome synapsis, that was followed by apoptotic cell death. Thus, murine Msh5 promotes synapsis of homologous chromosomes in meiotic prophase I.


Subject(s)
Meiosis/physiology , Proteins/physiology , Animals , Base Pair Mismatch , Cell Cycle Proteins , Cell Line , DNA-Binding Proteins , Female , Infertility, Female/etiology , Infertility, Male/etiology , Male , Mice , Mice, Inbred C57BL , Oocytes , Proteins/genetics , Spermatocytes/physiology , Synaptonemal Complex/physiology
13.
Proc Natl Acad Sci U S A ; 96(7): 3911-5, 1999 Mar 30.
Article in English | MEDLINE | ID: mdl-10097137

ABSTRACT

Deficiency in genes involved in DNA mismatch repair increases susceptibility to cancer, particularly of the colorectal epithelium. Using Msh2 null mice, we demonstrate that this genetic defect renders normal intestinal epithelial cells susceptible to mutation in vivo at the Dlb-1 locus. Compared with wild-type mice, Msh2-deficient animals had higher basal levels of mutation and were more sensitive to the mutagenic effects of temozolomide. Experiments using Msh2-deficient cells in vitro suggest that an element of this effect is attributable to increased clonogenicity. Indeed, we show that Msh2 plays a role in the in vivo initiation of apoptosis after treatment with temozolomide, N-methyl-N'-nitro-N-nitrosoguanidine, and cisplatin. This was not influenced by the in vivo depletion of O6-alkylguanine-DNA-alkyltransferase after administration of O6-benzylguanine. By analyzing mice mutant for both Msh2 and p53, we found that the Msh2-dependent apoptotic response was primarily mediated through a p53-dependent pathway. Msh2 also was required to signal delayed p53-independent death. Taken together, these studies characterize an in vivo Msh2-dependent apoptotic response to methylating agents and raise the possibility that Msh2 deficiency may predispose to malignancy not only through failed repair of mismatch DNA lesions but also through the failure to engage apoptosis.


Subject(s)
Apoptosis/genetics , DNA Repair , DNA-Binding Proteins , Dacarbazine/analogs & derivatives , Intestinal Mucosa/drug effects , Intestine, Small/drug effects , Mutagenesis , Mutagens/toxicity , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Animals , Apoptosis/drug effects , Cells, Cultured , Cisplatin/toxicity , Dacarbazine/toxicity , Guanine/analogs & derivatives , Guanine/toxicity , Intestinal Mucosa/cytology , Intestinal Mucosa/pathology , Intestine, Small/cytology , Intestine, Small/pathology , Methylnitronitrosoguanidine/toxicity , Mice , Mice, Inbred BALB C , Mice, Inbred Strains , Mice, Knockout , MutS Homolog 2 Protein , Proto-Oncogene Proteins/deficiency , Stem Cells/cytology , Stem Cells/physiology , Temozolomide
14.
Exp Cell Res ; 246(1): 1-10, 1999 Jan 10.
Article in English | MEDLINE | ID: mdl-9882509

ABSTRACT

The majority of tumors associated with the nonpolyposis form of familial colorectal cancer (HNPCC) shows a specific form of genetic instability which is manifested by length alterations of mono- or dinucleotide repeat sequences [e.g., (A)n or (CA)n]. This phenomenon was termed the RER+ (replication error-positive) phenotype, MSI or MIN (microsatellite instability), and found to result from defects in the cells' DNA mismatch repair system. This system recognizes and restores misincorporated bases or slippage errors which frequently occur during DNA replication. Loss of DNA mismatch repair therefore strongly accelerates the evolutionary process of mutagenesis and selection which underlies the development of cancer. In addition to mutation avoidance, DNA mismatch repair also plays a crucial role in the toxicity of a number of DNA-damaging drugs that are used in cancer chemotherapy. In experimental systems, mismatch-repair-deficient cells are highly tolerant to the methylating chemotherapeutic drugs streptozocin and temozolomide and, albeit to a lesser extent, to cisplatin and doxorubicin. These drugs are therefore expected to be less effective on mismatch-repair-deficient tumors in humans. MIN was also found in a substantial portion of sporadic (nonfamilial) human tumors. However, in many cases the extent of microsatellite instability was not as dramatic as found in HNPCC-related tumors and the underlying genetic defect is unclear. Therefore, while the mismatch repair status of tumors may become an important determinant in the choice of chemotherapeutic intervention, the significance of MIN in sporadic cancer remains elusive.


Subject(s)
Microsatellite Repeats/genetics , Neoplasms/genetics , Trinucleotide Repeat Expansion/genetics , Base Pair Mismatch/genetics , Cisplatin/pharmacology , DNA Methylation , DNA Repair/genetics , Genetic Markers , Humans , Neoplasms/drug therapy , Prognosis
15.
Proc Natl Acad Sci U S A ; 95(20): 11915-20, 1998 Sep 29.
Article in English | MEDLINE | ID: mdl-9751765

ABSTRACT

Chronic oxidative stress may play a critical role in the pathogenesis of many human cancers. Here, we report that mouse embryonic stem (ES) cells deficient in DNA mismatch repair responded abnormally when exposed to low levels of ionizing radiation, a stress known to generate oxidative DNA damage. ES cells derived from mice carrying either one or two disrupted Msh2 alleles displayed an increased survival following protracted exposures to low-level ionizing radiation as compared with wild-type ES cells. The increases in survival exhibited by ES cells deficient in DNA mismatch repair appeared to have resulted from a failure to efficiently execute cell death (apoptosis) in response to radiation exposure. For each of the ES cell types, prolonged low-level radiation treatment generated oxidative genome damage that manifested as an accumulation of oxidized bases in genomic DNA. However, ES cells from Msh2(+/-) and Msh2(-/-) mice accumulated more oxidized bases as a consequence of low-level radiation exposure than ES cells from Msh2(+/+) mice. The propensity for normal cells with mismatch repair enzyme deficiencies, including cells heterozygous for inactivating mismatch repair enzyme gene mutations, to survive promutagenic genome insults accompanying oxidative stresses may contribute to the increased cancer risk characteristic of the hereditary nonpolyposis colorectal cancer syndrome.


Subject(s)
DNA-Binding Proteins , Mutation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Alleles , Animals , Apoptosis/genetics , Apoptosis/radiation effects , Cell Line , Colorectal Neoplasms, Hereditary Nonpolyposis/etiology , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , DNA Damage , DNA Repair/genetics , Heterozygote , Humans , Mice , MutS Homolog 2 Protein , Oxidative Stress/genetics , Oxidative Stress/radiation effects , Stem Cells/drug effects , Stem Cells/metabolism , Stem Cells/radiation effects , Thioguanine/pharmacology
16.
Oncogene ; 17(1): 1-12, 1998 Jul 09.
Article in English | MEDLINE | ID: mdl-9671308

ABSTRACT

The yeast-derived Flp-frt site-specific DNA recombination system was used to achieve pituitary-specific inactivation of the retinoblastoma (Rb) tumor suppressor gene. Whereas mice carrying only frt sites in both alleles of Rb remain tumor free, tumorigenesis ensues when the Flp recombinase is expressed. The rate of tumorigenesis in these mice depends both on the expression level of the Flp recombinase and on the presence of frt sites in one or both Rb alleles. This permitted a more accurate definition of the consecutive steps in pituitary tumorigenesis. Our study illustrates the potential of this approach for studying sporadic cancer in a defined mouse model.


Subject(s)
DNA Nucleotidyltransferases/metabolism , Gene Expression Regulation, Neoplastic , Genes, Retinoblastoma , Pituitary Neoplasms/genetics , Alleles , Animals , Apoptosis , Cell Division , Cell Line , Disease Progression , Female , Gene Deletion , Male , Mice , Mice, Transgenic , Pituitary Gland/growth & development , Pituitary Gland/pathology , Pituitary Neoplasms/pathology , Rabbits , Rats
17.
J Exp Med ; 187(11): 1735-43, 1998 Jun 01.
Article in English | MEDLINE | ID: mdl-9607915

ABSTRACT

To investigate the possible involvement of DNA repair in the process of somatic hypermutation of rearranged immunoglobulin variable (V) region genes, we have analyzed the occurrence, frequency, distribution, and pattern of mutations in rearranged Vlambda1 light chain genes from naive and memory B cells in DNA repair-deficient mutant mouse strains. Hypermutation was found unaffected in mice carrying mutations in either of the following DNA repair genes: xeroderma pigmentosum complementation group (XP)A and XPD, Cockayne syndrome complementation group B (CSB), mutS homologue 2 (MSH2), radiation sensitivity 54 (RAD54), poly (ADP-ribose) polymerase (PARP), and 3-alkyladenine DNA-glycosylase (AAG). These results indicate that both subpathways of nucleotide excision repair, global genome repair, and transcription-coupled repair are not required for somatic hypermutation. This appears also to be true for mismatch repair, RAD54-dependent double-strand-break repair, and AAG-mediated base excision repair.


Subject(s)
B-Lymphocytes/immunology , DNA Repair/physiology , Gene Rearrangement, B-Lymphocyte , Genes, Immunoglobulin , Immunologic Memory/immunology , Mutation , Animals , Immunoglobulin Variable Region/genetics , Immunoglobulin lambda-Chains/genetics , Mice , Mice, Mutant Strains , Polymerase Chain Reaction
18.
Genes Dev ; 12(11): 1599-609, 1998 Jun 01.
Article in English | MEDLINE | ID: mdl-9620848

ABSTRACT

Hemizygosity for the retinoblastoma gene RB in man strongly predisposes to retinoblastoma. In the mouse, however, Rb hemizygosity leaves the retina normal, whereas in Rb-/- chimeras pRb-deficient retinoblasts undergo apoptosis. To test whether concomitant inactivation of the Rb-related gene p107 is required to unleash the oncogenic potential of pRb deficiency in the mouse retina, we inactivated both Rb and p107 by homologous recombination in embryonic stem cells and generated chimeric mice. Retinoblastomas were found in five out of seven adult pRb/p107-deficient chimeras. The retinal tumors showed amacrine cell differentiation, and therefore originated from cells committed to the inner but not the outer nuclear layer. Retinal lesions were already observed at embryonic day 17.5. At this stage, the primitive nuclear layer exhibited severe dysplasia, including rosette-like arrangements, and apoptosis. These findings provide formal proof for the role of loss of Rb in retinoblastoma development in the mouse and the first in vivo evidence that p107 can exert a tumor suppressor function.


Subject(s)
Neoplasms, Experimental/genetics , Nuclear Proteins/genetics , Retinal Neoplasms/genetics , Retinoblastoma Protein/genetics , Retinoblastoma/genetics , Animals , Apoptosis , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Humans , Mice , Mice, Knockout , Neoplasms, Experimental/pathology , Retinal Neoplasms/pathology , Retinoblastoma/pathology , Retinoblastoma-Like Protein p107
19.
Cancer Res ; 58(2): 248-55, 1998 Jan 15.
Article in English | MEDLINE | ID: mdl-9443401

ABSTRACT

Hemizygous germ-line defects in mismatch repair (MMR) genes underlie hereditary nonpolyposis colorectal cancer (HNPCC). Loss of the wild-type allele results in a mutator phenotype, accelerating tumorigenesis. Tumorigenesis specifically occurs in the gastrointestinal and genitourinary tracts; the cause of this tissue specificity is elusive. To understand the etiology and tissue distribution of tumors in HNPCC, we have developed mouse models carrying a deficiency in the MMR gene Msh2. Most of the completely Msh2-deficient mice succumbed to lymphomas at an early age; lymphomagenesis was synergistically enhanced by exposure to ethylnitrosourea. Lymphomas were absent in immunocompromised Tap1-/-;Msh2-/- mice; these mice generally succumbed to HNPCC-like tumors. Together, these data suggest that the HNPCC tumor spectrum is determined by exposure of MMR-deficient cells to exogenous mutagens, rather than by tissue-specific loss of the wild-type MMR allele or by immune surveillance. Msh2 hemizygous mice had an elevated tumor incidence that, surprisingly, was rarely correlated with loss of the Msh2+ allele. To develop a model for intestinal tumorigenesis in HNPCC, we introduced the Min allele of the Apc tumor suppressor gene. We observed loss of the wild-type Msh2 allele in a significant fraction of intestinal tumors in Apc+/Min;Msh2+/- mice. In some of the latter tumors, one area of the tumor displayed loss of the Msh2+ allele, but not of the Apc+ allele, whereas another area displayed the inverse genotype. This apparent biclonality might indicate a requirement for collaboration between independent tumor clones during intestinal tumorigenesis.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis/etiology , Colorectal Neoplasms, Hereditary Nonpolyposis/pathology , Disease Models, Animal , Adenomatous Polyposis Coli Protein , Animals , Clone Cells , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Cytoskeletal Proteins/genetics , DNA-Binding Proteins/genetics , Ethylnitrosourea/pharmacology , Female , Gene Deletion , Immunocompromised Host , Loss of Heterozygosity , Lymphoma/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , MutS Homolog 2 Protein , Proto-Oncogene Proteins/genetics , Survival Rate
20.
Mol Cell Biol ; 17(8): 4782-91, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9234734

ABSTRACT

The E2A gene products, E12 and E47, are critical for proper early B-cell development and commitment to the B-cell lineage. Here we reveal a new role for E2A in T-lymphocyte development. Loss of E2A activity results in a partial block at the earliest stage of T-lineage development. This early T-cell phenotype precedes the development of a T-cell lymphoma which occurs between 3 and 9 months of age. The thymomas are monoclonal and highly malignant and display a cell surface phenotype similar to that of immature thymocytes. In addition, the thymomas generally express high levels of c-myc. As assayed by comparative genomic hybridization, each of the tumor populations analyzed showed a nonrandom gain of chromosome 15, which contains the c-myc gene. Taken together, the data suggest that the E2A gene products play a role early in thymocyte development that is similar to their function in B-lineage determination. Furthermore, the lack of E2A results in development of T-cell malignancies, and we propose that E2A inactivation is a common feature of a wide variety of human T-cell proliferative disorders, including those involving the E2A heterodimeric partners tal-1 and lyl-1.


Subject(s)
DNA-Binding Proteins/physiology , Lymphoma, T-Cell/immunology , T-Lymphocytes/cytology , Thymus Gland/immunology , Thymus Neoplasms/immunology , Transcription Factors , Animals , Cell Differentiation , Cell Extracts , Cell Nucleus/metabolism , Chromosome Aberrations , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Genes, myc , Lymphocyte Subsets , Lymphoma, T-Cell/genetics , Mice , Mice, Knockout , Mice, Nude , TCF Transcription Factors , Thymoma/genetics , Thymoma/immunology , Thymus Gland/growth & development , Thymus Neoplasms/genetics , Transcription Factor 7-Like 1 Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...