Tumor regionality in the mouse intestine reflects the mechanism of loss of Apc function.

Inherited colorectal cancer syndromes in humans exhibit regional specificity for tumor formation. By using mice with germline mutations in the adenomatous polyposis coli gene (Apc) and/or DNA mismatch repair genes, we have analyzed the genetic control of tumor regionality in the mouse small intestine. In C57BL/6 mice heterozygous for the Apc multiple intestinal neoplasia mutation (Apc(Min)), in which tumors are initiated by loss of heterozygosity by means of somatic recombination, tumors form preferentially in the distal region of the small intestine. By contrast, the formation of tumors initiated by allelic silencing on the AKR Apc(Min) genetic background is strongly skewed toward the ileocecal junction. A third tumor regionality is displayed by tumors that develop in MMR-deficient Apc(Min/+) mice, in which mutation of the Apc gene is responsible for tumor initiation. Thus, tumor regionality in the small intestine of Apc(Min/+) reflects the mechanism by which the wild-type allele of Apc is inactivated. We have reexamined the mechanism of Apc loss in tumors from Apc(1638N/+) mice, in which tumors of the small intestine develop in a regional pattern overlapping that of mismatch repair-deficient mice. In contrast to previous reports, we find that tumors from Apc(1638N/+) mice on a congenic C57BL/6 background maintain the wild-type allele of Apc. Our studies demonstrate a pathway-specific regionality for tumor development in mouse models for inherited intestinal cancer, an observation that is reminiscent of the regional preference for tumor development in the human colon. Perhaps, the power of mouse genetics and biology can be harnessed to identify genetic and other factors that contribute to tumor regionality.

[1]  A. Rustgi Gastrointestinal cancers : biology, diagnosis, and therapy , 1995 .

[2]  K. Kinzler,et al.  Erratum: Multiple Intestinal Neoplasia Caused By a Mutation in the Murine Homolog of the APC Gene , 1992, Science.

[3]  R Fodde,et al.  A targeted chain-termination mutation in the mouse Apc gene results in multiple intestinal tumors. , 1994, Proceedings of the National Academy of Sciences of the United States of America.

[4]  Darryl Shibata,et al.  Tumour susceptibility and spontaneous mutation in mice deficient in Mlh1, Pms1 and Pms2 DMA mismatch repair , 1998, Nature Genetics.

[5]  J. Hornaday,et al.  Cancer Facts & Figures 2004 , 2004 .

[6]  Y. Hérault,et al.  Engineering chromosomes in mice through targeted meiotic recombination (TAMERE) , 1998, Nature Genetics.

[7]  K. Haigis,et al.  A Robertsonian translocation suppresses a somatic recombination pathway to loss of heterozygosity , 2003, Nature Genetics.

[8]  D. Ward,et al.  Mutation in the DNA mismatch repair gene homologue hMLH 1 is associated with hereditary non-polyposis colon cancer , 1994, Nature.

[9]  C. Boland,et al.  Genetics, natural history, tumor spectrum, and pathology of hereditary nonpolyposis colorectal cancer: an updated review. , 1993, Gastroenterology.

[10]  N. Copeland,et al.  The human mutator gene homolog MSH2 and its association with hereditary nonpolyposis colon cancer , 1993, Cell.

[11]  T. Prolla,et al.  Involvement of mouse Mlh1 in DNA mismatch repair and meiotic crossing over , 1996, Nature Genetics.

[12]  Hereditary nonpolyposis colorectal cancer. , 2000, Seminars in surgical oncology.

[13]  S. Dudley,et al.  Mlh1 deficiency enhances several phenotypes of ApcMin/+ mice , 2000, Oncogene.

[14]  R. Kucherlapati,et al.  Tumorigenesis in Mlh1 and Mlh1/Apc1638N mutant mice. , 1999, Cancer research.

[15]  J. Greer,et al.  Targeting genes for self-excision in the germ line. , 1999, Genes & development.

[16]  H. Pitot,et al.  A dominant mutation that predisposes to multiple intestinal neoplasia in the mouse. , 1990, Science.

[17]  A. Common Gastrointestinal Cancers: Biology, Diagnosis, and Therapy , 1996 .

[18]  C. Midgley,et al.  A resistant genetic background leading to incomplete penetrance of intestinal neoplasia and reduced loss of heterozygosity in ApcMin/+ mice. , 1998, Proceedings of the National Academy of Sciences of the United States of America.

[19]  K. Haigis,et al.  Intestinal adenomas can develop with a stable karyotype and stable microsatellites , 2002, Proceedings of the National Academy of Sciences of the United States of America.

[20]  A. Moser,et al.  Studies of neoplasia in the Min mouse. , 1997, Biochimica et biophysica acta.

[21]  C. F. Wu,et al.  Resampling Inference with Complex Survey Data , 1988 .

[22]  M. Radman,et al.  Inactivation of the mouse Msh2 gene results in mismatch repair deficiency, methylation tolerance, hyperrecombination, and predisposition to cancer , 1995, Cell.

[23]  E. Lander,et al.  Genetic identification of Mom-1, a major modifier locus affecting Min-induced intestinal neoplasia in the mouse , 1993, Cell.

[24]  R. Kucherlapati,et al.  Tumor-associated Apc mutations in Mlh1−/−Apc1638N mice reveal a mutational signature of Mlh1 deficiency , 2000, Oncogene.

[25]  R. Kucherlapati,et al.  Loss of Apc and the entire chromosome 18 but absence of mutations at the Ras and Tp53 genes in intestinal tumors from Apc1638N, a mouse model for Apc-driven carcinogenesis. , 1997, Carcinogenesis.

[26]  A. Moser,et al.  Loss of Apc+ in intestinal adenomas from Min mice. , 1994, Cancer research.

[27]  E. Lander,et al.  Mom1 is a semi-dominant modifier of intestinal adenoma size and multiplicity in Min/+ mice. , 1996, Genetics.

[28]  R. Fodde,et al.  Mechanisms of APC-driven tumorigenesis: lessons from mouse models , 1999, Cytogenetic and Genome Research.

[29]  H Cheng,et al.  Clonal analysis of mouse intestinal epithelial progenitors. , 1999, Gastroenterology.