Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Mamm Genome ; 12(8): 606-11, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11471054

ABSTRACT

B6.129S7-Gtrosa26 (ROSA26) mice carry a LacZ-neo(R) insertion on Chromosome (Chr) 6, made by promoter trapping with AB1 129 ES cells. Female C57BL/6J Apc(Min/+) (B6 Min/+) mice are very susceptible to the induction of mammary tumors after treatment with ethylnitrosourea (ENU). However, ENU-treated B6 mice carrying both Apc(Min) and ROSA26 are resistant to mammary tumor formation. Thus, ROSA26 mice carry a modifier of Min-induced mammary tumor susceptibility. We have previously mapped the modifier to a 4-cM interval of 129-derived DNA that also contains the ROSA26 insertion. Here we report additional evidence for the effect of the ROSA26 insertion on mammary tumor formation. To test the hypothesis that the resistance was due to a linked modifier locus, we utilized two approaches. We have derived and tested two lines of mice that are congenic for 129-derived DNA within the minimal modifier interval and show that they are as susceptible to mammary tumors as are B6 mice. Additionally, we analyzed a backcross population segregating for the insertion and show that mice carrying the insertion are more resistant to mammary tumor development than are mice not carrying the insertion. Thus, the resistance is not due to a 129-derived modifier allele, but must be due to the ROSA26 insertion. In addition, the effect of the ROSA26 insertion can be detected in a backcross population segregating for other mammary modifiers.


Subject(s)
Genes, APC , Lac Operon/genetics , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Proteins/genetics , Alleles , Animals , Animals, Congenic , Chromosome Mapping , Chromosomes/genetics , Crosses, Genetic , Ethylnitrosourea/pharmacology , Female , Gene Expression Regulation, Neoplastic , Genetic Markers/genetics , Incidence , Male , Mammary Neoplasms, Experimental/chemically induced , Mice , Mice, Inbred C57BL , Mutation/genetics , RNA, Untranslated
2.
Cancer Res ; 61(8): 3480-5, 2001 Apr 15.
Article in English | MEDLINE | ID: mdl-11309311

ABSTRACT

Treatment of female C57BL/6J (B6) mice carrying the mutant Min allele of the adenomatous polyposis coli (Apc) gene with ethylnitrosourea (ENU) results in approximately 90% of mice developing an average of three mammary tumors within 65 days. As a first step in the identification of loci modifying susceptibility to ENU-induced mammary tumors and hyperplasias, we have tested ENU-treated Apc(Min)/+ (Min/+) mice on several hybrid backgrounds for susceptibility to mammary and intestinal tumors. C57BR/cdJxB6 (BRB6) Min/+ mice were more sensitive to development of mammary squamous cell carcinomas than B6 Min/+ mice. In contrast, Min/+ hybrids between B6 and FVB/NTac (FVB), 129X1/SvJ (129X1), and 129S6/SvEvTac (129S6) were all significantly more resistant to mammary carcinoma development. However, mice from these three crosses developed more focal mammary hyperplasias than did the B6 or BRB6 Min/+ mice. Susceptibility to intestinal tumors was independent of mammary tumor susceptibility in most hybrids. These results indicate that genetic background can affect independently the phenotypes conferred by the Min allele of APC:


Subject(s)
Genes, APC/genetics , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/genetics , Alleles , Animals , Crosses, Genetic , Female , Genetic Predisposition to Disease , Hyperplasia/genetics , Intestinal Neoplasms/genetics , Male , Mice , Mice, Inbred C57BL
3.
Carcinogenesis ; 22(2): 343-6, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11181458

ABSTRACT

The proteins encoded by BRCA1 and ATM may be important in DNA repair and maintenance of genomic integrity. Women heterozygous for a mutation in BRCA1 have an increased incidence of breast cancer. Some evidence also suggests that female carriers of ATM mutations may be susceptible to breast cancer. However, mice carrying one mutant allele of Brca1 or ATM are not highly susceptible to breast cancer. We proposed that heterozygosity for a mutant allele of Brca1 or ATM may confer a decreased ability to repair DNA damage. Such a defect might lead to a heightened sensitivity to tumor development in susceptible animal models. Therefore, mice predispose to mammary tumor development might show an increased susceptibility if they also carry an ATM or Brca1 mutation. C57BL/6J (B6) MIN/+ mice are predisposed to mammary and intestinal tumors and exposure to the point mutagen ethylnitrosourea (ENU) markedly increases mammary tumor multiplicity and incidence. To test our hypothesis, B6.MIN/+ male mice were crossed with 129S6/SvEvTac females heterozygous for a mutant allele of either Brca1 or ATM. Female progeny from each cross were treated with ENU and followed for tumor development. Only MIN/+ F1 females developed mammary tumors and heterozygosity for a mutant Brca1 or ATM allele had no effect on mammary or intestinal tumor incidence and multiplicity. These results suggest that heterozygosity for a mutation in Brca1 or ATM: does not affect MIN-induced tumorigenesis in mice under these conditions. Additionally, exposure to a somatic point mutagen does not increase tumor development in mice carrying Brca1 or ATM mutations.


Subject(s)
Ethylnitrosourea/toxicity , Genes, BRCA1/genetics , Genetic Predisposition to Disease , Loss of Heterozygosity/genetics , Mammary Neoplasms, Experimental/genetics , Mutation , Protein Serine-Threonine Kinases/genetics , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins , DNA Primers/chemistry , DNA-Binding Proteins , Female , Incidence , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Knockout , Phenotype , Polymerase Chain Reaction , Tumor Suppressor Proteins
4.
Mamm Genome ; 11(12): 1058-62, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11130972

ABSTRACT

B6.129S7-Gtrosa26 (B6.R26) mice carry a LacZ-neoR insertion on Chromosome (Chr) 6, made by promoter trapping with 129 ES cells. Female C57BL/6J ApcMin/+ (B6Min/+) mice are highly susceptible to intestinal tumors and to the induction of mammary tumors after treatment with ethylnitrosourea (ENU). However, B6.R26/+ Min/+ females develop fewer mammary and intestinal tumors after ENU treatment than do B6 Min/+ mice. B6.R26/+ mice from two independently derived congenic lines show this modifier effect. Each of these congenic lines carries approximately 20 cM of 129-derived DNA flanking the insertion, raising the possibility that the resistance is due to a linked modifier locus. To further map the modifier locus, we have generated several lines of mice carrying different regions of the congenic interval. We have found that resistance to mammary and intestinal tumors in ENU-treated Min/+ mice maps to a minimum 4-cM interval that includes the ROSA26 LacZ-neoR insertion. Therefore, the resistance to tumor development is due to either the ROSA26 insertion or a very tightly linked modifier locus.


Subject(s)
Genetic Predisposition to Disease , Intestinal Neoplasms/genetics , Mammary Neoplasms, Experimental/genetics , Animals , Base Sequence , DNA Primers , Ethylnitrosourea/toxicity , Female , Genes, APC , Male , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Mutant Strains
5.
Proc Natl Acad Sci U S A ; 97(7): 3461-6, 2000 Mar 28.
Article in English | MEDLINE | ID: mdl-10716720

ABSTRACT

The interaction between mutations in the tumor-suppressor genes Apc and p53 was studied in congenic mouse strains to minimize the influence of polymorphic modifiers. The multiplicity and invasiveness of intestinal adenomas of Apc(Min/+) (Min) mice was enhanced by deficiency for p53. In addition, the occurrence of desmoid fibromas was strongly enhanced by p53 deficiency. The genetic modifier Mom1 and the pharmacological agents piroxicam and difluoromethylornithine each reduced intestinal adenoma multiplicity in the absence of p53 function. Mom1 showed no influence on the development of desmoid fibromas, whereas the combination of piroxicam and difluoromethylornithine exerted a moderate effect. The ensemble of tumor suppressors and modifiers of a neoplastic process can be usefully analyzed in respect to tissue specificity and synergy.


Subject(s)
Adenoma/genetics , Intestinal Neoplasms/genetics , Neoplasms, Multiple Primary/genetics , Adenoma/pathology , Animals , Eflornithine/pharmacology , Female , Fibromatosis, Aggressive/genetics , Genes, APC , Genes, p53 , Intestinal Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Neoplasms, Multiple Primary/pathology , Piroxicam/pharmacology
6.
Proc Natl Acad Sci U S A ; 95(18): 10826-31, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9724789

ABSTRACT

Previous studies of Min/+ (multiple intestinal neoplasia) mice on a sensitive genetic background, C57BL/6 (B6), showed that adenomas have lost heterozygosity for the germ-line ApcMin mutation in the Apc (adenomatous polyposis coli) gene. We now report that on a strongly resistant genetic background, AKR/J (AKR), Min-induced adenoma multiplicity is reduced by about two orders of magnitude compared with that observed on the B6 background. Somatic treatment with a strong mutagen increases tumor number in AKR Min/+ mice in an age-dependent manner, similar to results previously reported for B6 Min/+ mice. Immunohistochemical analyses indicate that Apc expression is suppressed in all intestinal tumors from both untreated and treated AKR Min/+ mice. However, the mechanism of Apc inactivation in AKR Min/+ mice often differs from that observed for B6 Min/+ mice. Although loss of heterozygosity is observed in some tumors, a significant percentage of tumors showed neither loss of heterozygosity nor Apc truncation mutations. These results extend our understanding of the effects of genetic background on Min-induced tumorigenesis in several ways. First, the AKR strain carries modifiers of Min in addition to Mom1. This combination of AKR modifiers can almost completely suppress spontaneous intestinal tumorigenesis associated with the Min mutation. Second, even on such a highly resistant genetic background, tumor formation continues to involve an absence of Apc function. The means by which Apc function is inactivated is affected by genetic background. Possible scenarios are discussed.


Subject(s)
Adenoma/pathology , Genes, APC , Intestinal Neoplasms/pathology , Loss of Heterozygosity , Adenoma/chemically induced , Adenoma/genetics , Alleles , Animals , Carcinogens , Ethylnitrosourea , Germ-Line Mutation , Intestinal Neoplasms/chemically induced , Intestinal Neoplasms/genetics , Mice
7.
Cancer Res ; 57(10): 1999-2006, 1997 May 15.
Article in English | MEDLINE | ID: mdl-9157997

ABSTRACT

We have demonstrated previously that intestinal tumor formation in B6 Min/+ mice is always accompanied by loss of the wild-type adenomatous polyoposis coli (Apc) allele and that intestinal tumor multiplicity in B6 Min/+ mice can be significantly increased by treatment with a single dose of N-ethyl-N-nitrosourea (ENU). Here, we show that some tumors from ENU-treated Min/+ mice can form without complete elimination of Apc+. At least 25% of these tumors acquired somatic Apc truncation mutations. Interestingly, some ENU-induced tumors demonstrated loss of the Apc+ allelic marker examined by the quantitative PCR assay used here. Using two methods of mutation detection, we identified no Apc mutations in at least 12% of the tumors from ENU-treated B6 Min/+ mice. Finally, no H- or K-ras-activating mutations were detected in intestinal tumors from either untreated or ENU-treated Min/+ mice. The majority of somatic human APC mutations in intestinal tumors lead to APC truncation. Our results demonstrate that somatic Apc truncation mutations also frequently occur in ENU-induced intestinal tumors in Min mice.


Subject(s)
Intestinal Neoplasms/genetics , Mutation , Alleles , Animals , Carcinogens , Ethylnitrosourea , Female , Gene Deletion , Gene Expression Regulation, Neoplastic , Genes, APC , Genes, ras , Intestinal Neoplasms/chemically induced , Male , Mice , Mice, Inbred Strains
9.
Cancer Res ; 57(5): 812-4, 1997 Mar 01.
Article in English | MEDLINE | ID: mdl-9041176

ABSTRACT

We have tested the hypothesis that enteric bacteria are necessary for formation of intestinal adenomas in C57BL/6-ApcMin/+ mouse. Germ-free mice developed 2-fold fewer adenomas than conventional controls in the medial small intestine (7.3 versus 14.9; P < 0.003), but there were no significant differences in the rest of the intestinal tract. We conclude that microbial status does not strongly alter the adenoma phenotype in this mouse model of familial adenomatous polyposis. In parallel, we have found that C57BL/6-ApcMin/+ mice mutated at the beige locus, which controls natural killer activity, are also unaltered in adenoma multiplicity.


Subject(s)
Adenomatous Polyposis Coli/genetics , Intestinal Neoplasms/etiology , Intestines/microbiology , Killer Cells, Natural/immunology , Animals , Germ-Free Life , Intestine, Small/microbiology , Intestine, Small/pathology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains
10.
Genetics ; 144(4): 1777-85, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8978063

ABSTRACT

As genetic mapping of quantitative trait loci (QTL) becomes routine, the challenge is to identify the underlying genes. This paper develops rigorous genetic tests for evaluation of candidate genes for a QTL, involving determination of allelic status in inbred strains and fine-structure genetic mapping. For the Mom1 modifier of intestinal adenomas caused by ApcMin, these tests are used to evaluate two candidate genes: Pla2g2a, a secretory phospholipase, and Rap1GAP, a GTPase activating protein. Rap1GAP passes the first test but is excluded by a single fine-structure recombinant. Pla2g2a passes both tests and is a strong candidate for Mom1. Significantly, we also find that ApcMin-induced adenomas remain heterozygous for the Mom1 region, consistent with Mom1 acting outside the tumor lineage and encoding a secreted product.


Subject(s)
Adenoma/genetics , Chromosome Mapping , Genes, APC , Intestinal Neoplasms/genetics , Alleles , Animals , Mice
11.
Carcinogenesis ; 17(8): 1757-60, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8761438

ABSTRACT

The Min mouse provides a genetically defined model for inherited and sporadic forms of human colorectal tumorigenesis. To test the suitability of this model for the evaluation and optimization of chemopreventive agents, we examined the effects of sulindac on tumorigenesis in Min mice as this compound can inhibit colorectal tumorigenesis in human familial adenomatous polyposis patients. Treatment of Min mice with sulindac in their drinking water (84 mg/l) or diet (167 and 334 p.p.m.) resulted in a significantly decreased average tumor load. The conservation of sulindac activity in the Min mouse provides an opportunity to explore the mechanism of sulindac suppression as well as to test other potential chemopreventive agents.


Subject(s)
Anticarcinogenic Agents/pharmacology , Colorectal Neoplasms/prevention & control , Sulindac/pharmacology , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/pathology , Animals , Colorectal Neoplasms/genetics , Disease Models, Animal , Humans , Mice , Mice, Mutant Strains
12.
Cancer Res ; 56(4): 710-4, 1996 Feb 15.
Article in English | MEDLINE | ID: mdl-8631000

ABSTRACT

C57BL/6J-Min/+mice (n = 56), heterozygous for a nonsense mutation in the Apc gene, were randomized at weaning to seven groups, including groups treated with piroxicam at 0, 50, 100, and 200 ppm in the AIN93G diet. After only 6 weeks of treatment, intestinal adenomas and aberrant crypt foci were counted, and serum levels of piroxicam and thromboxane B2 were quantitated. Tumor multiplicity was decreased in a dose-dependent manner from 17.3 +/- 2.7 in the control to 2.1 +/- 1.1 (12%) in the high-dose piroxicam group (P < 0.001). Thromboxane B2 levels in plasma also decreased monotonically in parallel to the decrease in tumor multiplicity, consistent with the prostaglandin inhibitory effect of piroxicam. The Min mouse model demonstrates that the nonsteroidal anti-inflammatory drug piroxicam has strong biological and therapeutic effects, potentially useful for prevention of the early adenoma stage of tumor development.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anticarcinogenic Agents/pharmacology , Colonic Neoplasms/prevention & control , Genes, APC , Intestinal Neoplasms/prevention & control , Piroxicam/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/blood , Anticarcinogenic Agents/blood , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Crosses, Genetic , Dose-Response Relationship, Drug , Female , Heterozygote , Intestinal Neoplasms/genetics , Intestinal Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mutation , Piroxicam/blood , Random Allocation , Thromboxane B2/blood
13.
Cancer Res ; 55(19): 4479-85, 1995 Oct 01.
Article in English | MEDLINE | ID: mdl-7671263

ABSTRACT

The timing of intestinal tumor initiation in B6-Min/+ mice has been examined by treating mice at 5-35 days of age with a single i.p. injection of the direct-acting alkylating agent N-ethyl-N-nitrosourea (ENU). Treatment of Min/+ mice at 5-14 days of age resulted in a 3.8-fold increase in intestinal tumor multiplicity over untreated mice. Mice treated at 20-35 days of age showed only a 1.6-fold increase in tumor number. These results, in conjunction with examination of tumor multiplicities of untreated Min/+ mice as a function of age, suggest that the majority of intestinal tumors in Min/+ mice are initiated relatively early in life. Min/+ mice treated with ENU also showed an increase in the number of cystic intestinal crypts. However, the relationship between age at ENU treatment and cystic crypt multiplicity was distinct from that seen for intestinal adenomas. Mice treated at 5-9 days of age showed only a 1.9-fold increase in cystic crypts over untreated animals. By contrast, the increase in average cystic crypt multiplicity for mice treated at 10-35 days of age was 4.5-fold. In addition, 60% of Min/+ mice treated with ENU before 25 days of age developed epidermoid cysts, an extracolonic manifestation commonly associated with familial adenomatous polyposis in humans.


Subject(s)
Adenoma/chemically induced , Carcinogens/toxicity , Cysts/chemically induced , Epidermal Cyst/chemically induced , Ethylnitrosourea/toxicity , Intestinal Diseases/chemically induced , Intestinal Neoplasms/chemically induced , Skin Neoplasms/chemically induced , Age Factors , Animals , Female , Male , Mice
14.
Dev Dyn ; 203(4): 422-33, 1995 Aug.
Article in English | MEDLINE | ID: mdl-7496034

ABSTRACT

Mutation of the APC (adenomatous polyposis coli) gene is an early event in colon tumor development in humans. Mice carrying Min (multiple intestinal neoplasia), a mutant allele of Apc, develop intestinal and mammary tumors as adults. To study the role of the Apc gene in development, we have investigated the phenotype of embryos homozygous for ApcMin (Min). Development of the primitive ectoderm fails prior to gastrulation in homozygous Min embryos. By midgestation, the presumed homozygotes consist of a mass of trophoblast giant cells with an additional cluster of much smaller embryonic cells. These results indicate that functional Apc is required for normal growth of inner cell mass derivatives.


Subject(s)
Gastrula/physiology , Genes, APC/genetics , Mice, Inbred Strains/embryology , Alleles , Animals , Embryo, Mammalian/abnormalities , Embryo, Mammalian/physiology , Genes, Lethal , Genotype , Heterozygote , Homozygote , Mice , Mutation , Phenotype
15.
Eur J Cancer ; 31A(7-8): 1061-4, 1995.
Article in English | MEDLINE | ID: mdl-7576992

ABSTRACT

Min (multiple intestinal neoplasia) is a mutant allele of the murine Apc (adenomatous polyposis coli) locus, encoding a nonsense mutation at codon 850. Like humans with germline mutations in APC, Min/+ mice are predisposed to intestinal adenoma formation. The number of adenomas is influenced by modifier loci carried by different inbred strains. One modifier locus, Mom-1 (modifier of Min-1), maps to distal chromosome 4. Intestinal tumours from both B6 (C57BL/6J) and hybrid Min/+ mice show extensive loss of the wild-type allele at Apc. B6 Min/+ female mice are predisposed to spontaneous mammary tumours. The incidence of both intestinal and mammary tumours can be increased in an age-specific manner by treatment with ethylnitrosourea (ENU). Min mice provide a good animal model for studying the role of Apc and interacting genes in the initiation and progression of intestinal and mammary tumorigenesis.


Subject(s)
Adenomatous Polyposis Coli/genetics , Disease Models, Animal , Genes, APC , Mammary Neoplasms, Experimental/genetics , Animals , Female , Germ-Line Mutation , Mice , Mice, Inbred Strains
16.
Cancer Surv ; 25: 335-55, 1995.
Article in English | MEDLINE | ID: mdl-8718526

ABSTRACT

Mutation of the APC gene may be a common denominator of all human colon cancer--polypoid and non-polypoid familial cancer as well as sporadic occurrences. Fearon and Vogelstein (1990) have described a series of molecular changes during the progression of human colon cancer, beginning with mutations in APC. Min is a strain of the laboratory mouse carrying a nonsense mutation in Apc, the mouse homologue of APC. The Min strain has been used to test the effect of germline alterations in certain genes identified in the progression pathway of Fearon and Vogelstein. A deficiency in DNA cytosine methylase leads to a reduction in the tumour multiplicity of Min mice contrary to the a priori expectation based on the global hypomethylation of the DNA of early colonic neoplasms. Alterations in Kras had no perceptible effect on the tumour multiplicity of Min mice but may not have been successfully directed to the proliferative cell population. Constitutional mutation of p53 did not influence the multiplicity or histopathology of early Min induced intestinal tumours. The cause and effect analysis of the genetics of colon cancer is clearly in an early phase. An unlinked genetic factor interacting with Min in controlling intestinal tumour multiplicity is Mom1. A central goal for the near future is to identify the Mom1 gene product and to identify other loci that can interact with the Min mutation and affect tumour multiplicity or progression. Mouse chimaeras will permit an analysis of the clonality and cell autonomy of Min induced neoplasms and also of the action of Mom1. The results of these analyses will inform investigators as to what modes of prevention and therapy might be designed for particular tumour types. The Min strain thereby presents an opportunity to discover protective factors against human colon cancer.


Subject(s)
Adenomatous Polyposis Coli/genetics , Colorectal Neoplasms/genetics , Genes, APC , Intestinal Neoplasms/genetics , Alleles , Animals , Genes, p53 , Heterozygote , Homozygote , Humans , Mice , Neoplasms, Experimental/genetics , Phenotype
17.
Cancer Res ; 54(22): 5947-52, 1994 Nov 15.
Article in English | MEDLINE | ID: mdl-7954427

ABSTRACT

Allelic loss at the Apc locus in spontaneously occurring intestinal adenomas from mice heterozygous for the ApcMin nonsense mutation was analyzed using a site-specific quantitative polymerase chain reaction assay. All 97 of the intestinal adenomas analyzed showed extensive loss of the wild-type Apc (Apc+) allele. Quantitative polymerase chain reaction analysis of loci linked to Apc indicated loss of the chromosome carrying Apc+. Only one copy of the homologue carrying ApcMin remained in the intestinal adenomas. Possible reasons for the difference in the mechanism of Apc+ loss between human and Min mouse intestinal adenomas are discussed.


Subject(s)
Adenoma/genetics , Gene Deletion , Genes, APC/genetics , Intestinal Neoplasms/genetics , Animals , Base Sequence , Chromosome Mapping , Colonic Neoplasms/genetics , DNA Probes/genetics , Female , Intestine, Small , Male , Mice , Mice, Inbred AKR , Molecular Sequence Data , Polymerase Chain Reaction
19.
J Cell Biol ; 123(4): 877-93, 1993 Nov.
Article in English | MEDLINE | ID: mdl-8227147

ABSTRACT

SV-40 T antigen (TAg), human K-rasVal12, and a dominant negative mutant of human p53 (p53Ala143) have been expressed singly and in all possible combinations in postmitotic enterocytes distributed throughout the duodenal-colonic axis of 1-12-mo-old FVB/N transgenic mice to assess the susceptibility of this lineage to gene products implicated in the pathogenesis of human gut neoplasia. SV-40 TAg produces re-entry into the cell cycle. Transgenic pedigrees that produce K-rasVal12 alone, p53Ala143 alone, or K-rasVal12 and p53Ala143 have no detectable phenotypic abnormalities. However, K-rasVal12 cooperates with SV-40 TAg to generate marked proliferative and dysplastic changes in the intestinal epithelium. These abnormalities do not progress to form adenomas or adenocarcinomas over a 9-12-mo period despite sustained expression of the transgenes. Addition of p53Ala143 to enterocytes that synthesize SV-40 TAg and K-rasVal12 does not produce any further changes in proliferation or differentiation. Mice that carry one, two, or three of these transgenes were crossed to animals that carry Min, a fully penetrant, dominant mutation of the Apc gene associated with the development of multiple small intestinal and colonic adenomas. A modest (2-5-fold) increase in tumor number was noted in animals which express SV-40 TAg alone, SV-40 TAg and K-rasVal12, or SV-40 TAg, K-rasVal12 and p53Ala143. However, the histopathologic features of the adenomas were not altered and the gut epithelium located between tumors appeared similar to the epithelium of their single transgenic, bi-transgenic, or tri-transgenic parents without Min. These results suggest that (a) the failure of the dysplastic gut epithelium of SV-40 TAg X K-rasVal12 mice to undergo further progression to adenomas or adenocarcinomas is due to the remarkable protective effect of a continuously and rapidly renewing epithelium, (b) initiation of tumorigenesis in Min mice typically occurs in crypts rather than in villus-associated epithelial cell populations, and (c) transgenic mouse models of neoplasia involving members of the enterocytic lineage may require that gene products implicated in tumorigenesis be directed to crypt stem cells or their immediate descendants. Nonetheless, directing K-rasVal12 production to proliferating and nonproliferating cells in the lower and upper half of small intestinal and colonic crypts does not result in any detectable abnormalities.


Subject(s)
Disease Models, Animal , Intestinal Neoplasms/genetics , Neoplasm Proteins , Nerve Tissue Proteins , Analysis of Variance , Animals , Antigens, Polyomavirus Transforming/genetics , Antigens, Polyomavirus Transforming/metabolism , Base Sequence , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Differentiation , Cell Division , DNA , Fatty Acid-Binding Protein 7 , Fatty Acid-Binding Proteins , Genes, ras , Intestinal Mucosa/metabolism , Intestines/cytology , Mice , Mice, Transgenic , Molecular Sequence Data , Oncogenes , Phenotype , Restriction Mapping , Stem Cells/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
20.
Cell ; 75(4): 631-9, 1993 Nov 19.
Article in English | MEDLINE | ID: mdl-8242739

ABSTRACT

Mutations in the human APC gene caused various familial colon cancer syndromes. The Multiple intestinal neoplasia (Min) mouse provides an excellent model for familial colon cancer: it carries a mutant mouse Apc gene and develops many intestinal adenomas. Here, we analyze how this tumor phenotype is dramatically modified by genetic background. We report the genetic mapping of a locus that strongly modifies tumor number in Min/+ animals. This gene, Mom-1 (Modifier of Min-1), maps to distal chromosome 4 and controls about 50% of genetic variation in tumor number in two intraspecific backcrosses. The mapping is supported by a LOD score exceeding 14. Interestingly, Mom-1 lies in a region of synteny conservation with human chromosome 1p35-36, a region of frequent somatic loss of heterozygosity in a variety of human tumors, including colon tumors. These results provide evidence of a major modifier affecting expression of an inherited cancer syndrome.


Subject(s)
Colonic Neoplasms/genetics , Genes, Tumor Suppressor , Mice/genetics , Adenomatous Polyposis Coli/genetics , Animals , Chromosome Mapping , Chromosomes, Human, Pair 1 , Genes, Dominant , Humans , Mice, Mutant Strains , Neoplasms, Experimental/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...