Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Nutr Cancer ; 64(3): 401-8, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22439692

ABSTRACT

Increased caloric intake has been associated with increased risk for cancer of the large intestine. We studied caloric intake effect on tumor formation in Apc1638( N/+ ) mice, a preclinical model for human familial adenomatous polyposis. Mice were fed a controlled AIN-76A diet or a new Western-style diet (NWD). Intestinal tumor development was evaluated after 6 mo of feeding 1) AIN-76A diet (fed ad libitum) vs. AIN-76A (caloric intake reduced 30%); 2) NWD (fed ad libitum) vs. NWD (caloric intake reduced 30%); and 3) AIN-76A (fed ad libitum) vs. NWD (paired-fed with NWD providing equal caloric intakes to AIN-76A). Intestinal tumor incidences were 78-100% with intergroup variation P > 0.05; however, tumor multiplicity responded differently to dietary treatment: 1) Tumor multiplicity was unchanged after AIN-76A (caloric intake reduced 30% vs. mice fed AIN-76A ad libitum); 2) tumor multiplicity was unchanged after NWD (caloric intake reduced 30% vs. NWD ad libitum); and 3) tumor multiplicity increased 130% after NWD was paired-fed with the same caloric intake as mice fed AIN-76A ad libitum (P < 0.05). Body weights showed no association with tumor development. Findings indicated modified nutrients in NWD were mainly responsible for increased tumors in mice fed NWD vs. AIN-76A in this preclinical mouse model for human FAP.


Subject(s)
Colonic Neoplasms/pathology , Diet , Energy Intake , Adenomatous Polyposis Coli/pathology , Animals , Apoptosis , Body Weight , Cell Transformation, Neoplastic , Colon/pathology , Disease Models, Animal , Female , Male , Mice , Mice, Knockout
2.
Gastroenterology ; 138(3): 993-1002.e1, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19931261

ABSTRACT

BACKGROUND & AIMS: Mutations in the DNA mismatch repair (MMR) gene MSH2 cause Lynch syndromes I and II and sporadic colorectal cancers. Msh2(null) mice predominantly develop lymphoma and do not accurately recapitulate the colorectal cancer phenotype. METHODS: We generated and examined mice with a conditional Msh2 disruption (Msh2(LoxP)), permitting tissue-specific gene inactivation. ECMsh2(LoxP/LoxP) mice carried an EIIa-Cre transgene, and VCMsh2(LoxP/LoxP) mice carried a Villin-Cre transgene. We combined the VCMsh2(LoxP) allele with either Msh2(Delta7null) (VCMsh2(LoxP/null)) or Msh2(G674D) mutations (VCMsh2(LoxP/G674D)) to create allelic phase mutants. These mice were given cisplatin or 5-fluorouracil/leucovorin and oxaliplatin (FOLFOX), and their tumors were measured by magnetic resonance imaging. RESULTS: Embryonic fibroblasts from ECMsh2(LoxP/LoxP) mice do not express MSH2 and are MMR deficient. Reverse transcription, polymerase chain reaction, and immunohistochemistry from VCMsh2(LoxP/LoxP) mice demonstrated specific loss of Msh2 messenger RNA and protein from epithelial cells of the intestinal tract. Microsatellite instability was observed in all VCMsh2 strains and limited to the intestinal mucosa. Resulting adenomas and adenocarcinomas had somatic truncation mutations to the adenomatous polyposis coli (Apc) gene. VCMsh2(LoxP/LoxP) mice did not develop lymphoma. Comparison of allelic phase tumors revealed significant differences in multiplicity and size. When treated with cisplatin or FOLFOX, tumor size was reduced in VCMsh2(LoxP/G674D) but not VCMsh2(LoxP/null) tumors. The apoptotic response to FOLFOX was partially sustained in the intestinal mucosa of VCMsh2(LoxP/G674D) animals. CONCLUSIONS: Msh2(LoxP/LoxP) mice in combination with appropriate Cre recombinase transgenes have excellent potential for preclinical modeling of Lynch syndrome, MMR-deficient tumors of other tissue types, and use in drug development.


Subject(s)
Adenocarcinoma/drug therapy , Adenoma/drug therapy , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cisplatin/pharmacology , Colorectal Neoplasms, Hereditary Nonpolyposis/drug therapy , Intestinal Neoplasms/drug therapy , Mice, Knockout , MutS Homolog 2 Protein/deficiency , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenoma/genetics , Adenoma/metabolism , Adenoma/pathology , Animals , Apoptosis/drug effects , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/metabolism , Colorectal Neoplasms, Hereditary Nonpolyposis/pathology , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Fluorouracil/pharmacology , Gene Expression Regulation, Neoplastic , Gene Silencing , Genes, APC , Genotype , Immunohistochemistry , Integrases/genetics , Intestinal Neoplasms/genetics , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Leucovorin/pharmacology , Magnetic Resonance Imaging , Mice , Mice, Inbred C57BL , Microfilament Proteins/genetics , Microsatellite Instability , MutS Homolog 2 Protein/genetics , Mutation , Organoplatinum Compounds/pharmacology , Phenotype , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Tumor Burden/drug effects
3.
Carcinogenesis ; 30(11): 1923-6, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19755659

ABSTRACT

We have previously reported that sulindac, a non-steroidal anti-inflammatory drug, inhibited tumor formation in the small intestine but increased tumors in the colon of Apc(Min/+) mice, a model of human familial adenomatous polyposis. To further explore intestinal regional responses, we studied effects of sulindac on additional gene-targeted mouse models of human intestinal tumorigenesis; these were (i) Apc(1638N/+) mouse (chain termination mutation in exon 15 of the Apc gene); (ii) Mlh1(+/-) mouse (DNA mismatch repair deficiency, a mouse model of human hereditary non-polyposis colorectal cancer) and (iii) double-heterozygous Mlh1(+/-)Apc(1638N/+) mutant mouse. Mice were fed AIN-76A control diet with or without 0.02% sulindac for 6 months. Intestinal regional tumor incidence, multiplicity, volume and degree of inflammation were used as end points. The results showed the following: (i) sulindac inhibited tumor development in the small intestine of Apc(1638N/+) mice; (ii) in contrast, sulindac increased tumors in the small intestine of Mlh1 mutant mice, a neoplastic effect which persisted in heterozygous compound Mlh1(+/-)Apc(1638N/+) mutant mice; (iii) sulindac increased tumors in the cecum of all mice regardless of genetic background; (iv) sulindac decreased inflammation in the small intestine of Apc(1638N/+) mice, but it increased inflammation in the small intestine of Mlh1(+/-) mice and Mlh1(+/-)Apc(1638N/+) mice and (v) sulindac enhanced inflammation in the cecum of all mutant mice. Findings indicate that the effects of sulindac in the intestine of these mutant mouse models are probably related to genetic background and appear to be associated with its inflammatory-inducing response.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Colorectal Neoplasms, Hereditary Nonpolyposis , Mutation , Nuclear Proteins/genetics , Sulindac/adverse effects , Sulindac/pharmacology , Adenomatous Polyposis Coli/chemically induced , Adenomatous Polyposis Coli/drug therapy , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/pathology , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antineoplastic Agents/therapeutic use , Cecum/drug effects , Cecum/pathology , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/chemically induced , Colorectal Neoplasms, Hereditary Nonpolyposis/drug therapy , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/pathology , Disease Models, Animal , Humans , Inflammation/chemically induced , Inflammation/genetics , Intestine, Small/drug effects , Intestine, Small/pathology , Mice , MutL Protein Homolog 1 , Sulindac/therapeutic use
4.
Carcinogenesis ; 30(1): 88-92, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19017685

ABSTRACT

We reported previously that a new Western-style diet (NWD) for 18 months, consisting of elevated lipids and decreased calcium, vitamin D and methyl-donor nutrients, induced colonic tumors in normal C57Bl/6 mice [Newmark, H.L. et al. (2001) A Western-style diet induces benign and malignant neoplasms in the colon of normal C57Bl/6 mice. Carcinogenesis, 22, 1871-1875], suggesting a new mouse model for human sporadic colon cancer. Here, we have extended this study during a longer feeding period of 2 years wherein tumor formation, tumor inhibition by addition of dietary calcium and vitamin D and their effects on gene expression were determined. We also similarly tested individual supplements of methyl donor (transfer) nutrients (folic acid, choline, methionine and dietary fiber), but these had no significant effect on colonic tumor incidence or multiplicity, whereas supplementation with combined calcium and vitamin D produced significant decrease in both colon tumor incidence and multiplicity, during 2 years of feeding. No visible colonic tumors were found at 6 months, very few at 12 months, more at 18 months and significantly at 24 months. In a related study of gene changes of the mouse colonic mucosa at 6 months of feeding taken from this study, long before any tumors were visibly detectable, indicated altered profiles of gene expression linked to later risk of dietary initiation of colon tumor formation. This type of early genetic altered profile, an indication of increased risk of later colonic tumor development, may become a useful tool for prediction of colon tumor risk while the colon grossly still appears histologically and physiologically normal.


Subject(s)
Calcium/administration & dosage , Colonic Neoplasms/etiology , Diet , Disease Models, Animal , Vitamin D/administration & dosage , Animals , Body Weight , Colonic Neoplasms/prevention & control , Female , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL
5.
Cancer Res ; 68(19): 7803-10, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18829535

ABSTRACT

A defined rodent "new Western diet" (NWD), which recapitulates intake levels of nutrients that are major dietary risk factors for human colon cancer, induced colonic tumors when fed to wild-type C57Bl/6 mice for 1.5 to 2 years from age 6 weeks (two-thirds of their life span). Colonic tumors were prevented by elevating dietary calcium and vitamin D(3) to levels comparable with upper levels consumed by humans, but tumorigenesis was not altered by similarly increasing folate, choline, methionine, or fiber, each of which was also at the lower levels in the NWD that are associated with risk for colon cancer. The NWD significantly altered profiles of gene expression in the flat colonic mucosa that exhibited heterogeneity among the mice, but unsupervised clustering of the data and novel statistical analyses showed reprogramming of colonic epithelial cells in the flat mucosa by the NWD was similar to that initiated by inheritance of a mutant Apc allele. The NWD also caused general down-regulation of genes encoding enzymes involved in lipid metabolism and the tricarboxylic acid cycle in colonic epithelial cells before tumor formation, which was prevented by the supplementation of the NWD with calcium and vitamin D(3) that prevented colon tumor development, demonstrating profound interaction among nutrients. This mouse model of dietary induction of colon cancer recapitulates levels and length of exposure to nutrients linked to relative risk for human sporadic colon cancer, which represents the etiology of >90% of colon cancer in the United States and other Western countries.


Subject(s)
Colonic Neoplasms/etiology , Diet/adverse effects , Disease Models, Animal , Mice , Animals , Cluster Analysis , Colonic Neoplasms/epidemiology , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Female , Gene Expression Profiling , Genes, APC , Incidence , Male , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Mucin-1/genetics , Oligonucleotide Array Sequence Analysis , Signal Transduction/genetics
6.
Proc Natl Acad Sci U S A ; 105(40): 15493-8, 2008 Oct 07.
Article in English | MEDLINE | ID: mdl-18832169

ABSTRACT

To examine the role of Rb1 in gastrointestinal (GI) tumors, we generated mice with an Apc(1638N) allele, Rb(tm2brn) floxed alleles, and a villin-cre transgene (RBVCA). These animals had exon 19 deleted from Rb1 throughout the GI tract. We have shown previously that Rb1 deficiency is insufficient for GI tumor initiation, with inactivation of an Apc allele capable of overcoming the insufficiency. In this study we demonstrate that RBVCA mice have reduced median survival because of an increase in tumor incidence and multiplicity in the cecum and the proximal colon. Large intestinal tumors are predominantly adenomas, whereas the tumors of the small intestine are a mixture of adenomas and adenocarcinomas. We find truncation mutations to the second Apc allele in tumors of both the large and small intestine. Expression profiles of duodenal and cecal tumors relative to each other show unique gene subsets up and down regulated. Substantial expression patterns compare to human colorectal cancer, including recapitulation of embryonic genes. Our results indicate that Rb1 has significant influence over tumor location in the GI tract, and that both cecal and duodenal tumors initiate through inactivation of Apc. Expression profile analysis indicates the two tumor types differentially regulate distinct sets of genes that are over-expressed in a majority of human colorectal carcinomas.


Subject(s)
Cecal Neoplasms/genetics , Colonic Neoplasms/genetics , Gastrointestinal Tract/metabolism , Genes, APC , Retinoblastoma Protein/physiology , Alleles , Animals , Cecal Neoplasms/metabolism , Cecum/metabolism , Cecum/pathology , Colon/metabolism , Colon/pathology , Colonic Neoplasms/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Disease Models, Animal , Gastrointestinal Tract/pathology , Gene Expression Profiling , Mice , Mice, Transgenic , Mutation , Retinoblastoma Protein/genetics
7.
J Nutr ; 138(9): 1658-63, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18716166

ABSTRACT

Both epidemiological and experimental findings have indicated that components of Western diets influence colonic tumorigenesis. Among dietary constituents, calcium and cholecalciferol have emerged as promising chemopreventive agents. We have demonstrated that a Western-style diet (WD) with low levels of calcium and cholecalciferol and high levels of (n-6) PUFA, increased the incidence of neoplasia in mouse intestine compared with a standard AIN-76A diet; models included wild-type mice and mice with targeted mutations. In the present study, adenomatous polyposis coli (Apc)(1638N/+) mice carrying a heterozygous Apc mutation were fed either an AIN-76A diet, a WD, or a WD supplemented with calcium and cholecalciferol (WD/Ca/VitD3). Diets were fed for 24 wk and effects on cellular and molecular events were assessed by performing immunohistochemistry in colonic epithelium along the crypt-to-surface continuum. Feeding WD to Apc(1638N/+) mice not only enhanced cyclin D1 expression in colonic epithelium compared with AIN-76A treatment as previously reported but also significantly increased the expression of the antiapoptotic protein B-cell lymphoma 2 (Bcl-2) concomitantly with a decrease in the proapoptotic Bcl2-associated X protein and the number of apoptotic epithelial cells. WD treatment enhanced mutant Apc-driven small intestinal carcinogenesis and also resulted in the formation of a small number of colonic adenomas (0.16 +/- 0.09; P < 0.05). By contrast, the WD/Ca/VitD3 diet reversed WD-induced growth, promoting changes in colonic epithelium. Importantly, Apc(1638N/+) mice fed the WD/Ca/VitD3 diet did not develop colonic tumors, further indicating that dietary calcium and cholecalciferol have a key role in the chemoprevention of colorectal neoplasia in this mouse model of human colon cancer.


Subject(s)
Adenomatous Polyposis Coli/prevention & control , Apoptosis/drug effects , Calcium, Dietary/pharmacology , Cholecalciferol/pharmacology , Cyclin D1/metabolism , Adenomatous Polyposis Coli/metabolism , Adenomatous Polyposis Coli/pathology , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli Protein/metabolism , Animals , Body Weight , Calcium, Dietary/therapeutic use , Carcinogenicity Tests , Cholecalciferol/therapeutic use , Colon/pathology , Cyclin D1/genetics , Diet , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic/drug effects , Male , Mice , Mutation , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Random Allocation , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
8.
J Med Food ; 11(2): 201-6, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18358070

ABSTRACT

We previously reported hyperproliferation and hyperplasia in C57Bl/6 mouse mammary gland after feeding a Western-style diet (WD); these findings decreased after supplementing WD with increased calcium and vitamin D(3). We now again fortified WD with increased calcium and vitamin D(3) from two sources: (1) a food source, calcium- and vitamin D(3)-enriched yogurt (WD(y) diet) or (2) adding calcium and vitamin D(3) directly to WD (WD(CaD) diet). After 6 months of feeding the number of mammary ducts was higher in mice consuming WD compared to WD(y) (216.0 vs. 202.7, P <.05) and WD(CaD) (216.0 vs. 194.9, P <.001). The percentage of small ducts increased in WD compared to AIN-76A controls (23.3% vs. 17.4%) but was lower in the WD(y) (17.1%) and WD(CaD) (14.8%) groups. WD mice had higher numbers of epithelial cells per duct than WD(y) (33.2 vs. 27.4, P <.001) and WD(CaD) (33.2 vs. 27.8, P <.001) mice, and AIN-76A-fed mice had higher numbers than WD(y) (31.1 vs. 27.4, P <.005) or WD(CaD) (31.1 vs. 27.8, P <.01) mice. Mitotic index was higher in WD than in WD(CaD) mice (0.0020 vs. 0.0009, P <.001). Thus, small mammary gland ductules and mitosis increased after feeding WD and decreased after supplementing the diets with increased calcium and vitamin D(3), administered either in a dairy food (yogurt) or directly as calcium carbonate plus vitamin D(3) in WD, suggesting further study of these nutrients for their possible relationship to breast cancer prevention.


Subject(s)
Calcium/administration & dosage , Diet , Food, Fortified/analysis , Mammary Glands, Animal/drug effects , Vitamin D/administration & dosage , Animals , Apoptosis/drug effects , Cell Count , Epithelial Cells , Female , Mammary Glands, Animal/cytology , Mice , Mice, Inbred C57BL , Mitosis/drug effects
9.
J Med Food ; 10(1): 11-7, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17472461

ABSTRACT

Orange peel is a rich source of flavonoids with polymethoxyflavones as major constituents, compounds associated with potential antioxidant, anti-inflammatory, and antitumor activities. We studied the effect of an orange peel extract (OPE) on intestinal tumor growth in Apc(Min/+) mice, a mouse model for human familial adenomatous polyposis (FAP). The OPE contained 30% polymethoxyflavones, a mixture that included tangeretin (19.0%), heptamethoxyflavone (15.24%), tetramethoxyflavone (13.6%), nobiletin (12.49%), hexamethoxyflavone (11.06%), and sinensitin (9.16%). Apc(Min/+) mice were fed one of four diets: (1) AIN-76A control diet; (2) a new Western-style diet (NWD), i.e., AIN-76A diet modified with decreased calcium, vitamin D, and methyl-donor nutrients and increased lipid content); (3) NWD with 0.25% OPE; and (4) NWD with 0.5% OPE, with all additives premixed in the diet. After 9 weeks of feeding NWD to the Apc(Min/+) mice, tumors increased mainly in the colon, with tumor multiplicity increasing 5.3-fold and tumor volume increasing 6.7-fold. After feeding 0.5% OPE in NWD, the development of tumors markedly decreased, with multiplicity decreasing 49% in the small intestine and 38% in the colon. NWD also led to increased apoptosis in intestinal tumors, and 0.5% OPE in NWD further increased apoptosis in tumors of the small and large intestine. Findings indicated that OPE inhibited tumorigenesis in this preclinical mouse model of FAP, and increased apoptosis may have contributed to this effect.


Subject(s)
Anticarcinogenic Agents/administration & dosage , Citrus sinensis/chemistry , Fruit/chemistry , Intestinal Neoplasms/prevention & control , Plant Extracts/administration & dosage , Adenomatous Polyposis Coli/genetics , Animals , Apoptosis , Colonic Neoplasms/pathology , Colonic Neoplasms/prevention & control , Diet , Intestinal Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Phytotherapy
10.
J Med Food ; 10(1): 18-24, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17472462

ABSTRACT

Cancer chemoprevention via the ingestion of natural substances is a current topic of considerable interest. Flavonoids are a family of biologically active phytochemicals having a variety of biological effects. Orange peel extract (OPE) is an abundant source of polymethoxyflavones (PMFs) with potential chemopreventive properties. The OPE used here was a mixture containing tangeretin (19.0%), heptamethoxyflavone (15.24%), tetramethoxyflavone (13.6%), nobiletin (12.49%), hexamethoxyflavone (11.06%), and sinensitin (9.16%). C57Bl/6 mice were fed a new "Western-style" diet (NWD), which had previously induced atypical hyperplasias in mammary gland, and NWD supplemented with a standardized OPE containing 30% PMFs. Mice were fed one of four diets: (1) AIN-76A diet (control); (2) NWD; 0.25% OPE in NWD; or (4) 0.5% OPE in NWD. After 3 months of feeding, atypical hyperplasias developed in mammary glands of mice fed NWD, but not in controls. After feeding OPE in NWD, atypical hyperplasias per mouse decreased in frequency compared to feeding NWD alone (P < .05 in mice fed 0.25% OPE). Apoptosis increased in OPE-treated groups (P < .01) with no inhibition of mitosis. Thus, a standardized preparation of OPE with 30% PMFs decreased development of an atypical hyperplastic lesion and increased apoptosis in ductal epithelial cells of mouse mammary gland.


Subject(s)
Anticarcinogenic Agents/administration & dosage , Citrus sinensis/chemistry , Fruit/chemistry , Mammary Glands, Animal/pathology , Plant Extracts/administration & dosage , Animals , Apoptosis , Diet , Female , Flavones/administration & dosage , Flavones/analysis , Hyperplasia/prevention & control , Mice , Mice, Inbred C57BL , Mitosis , Phytotherapy , Plant Extracts/chemistry
11.
Anticancer Res ; 27(6B): 3807-12, 2007.
Article in English | MEDLINE | ID: mdl-18225536

ABSTRACT

BACKGROUND: The present study examines the effect of piroxicam, a non-steroidal anti-inflammatory drug, on tumor development in Mlh1+/- /Apc1638(N/+) mice, in a preclinical model of human colon cancer. MATERIALS AND METHODS: Mice were fed AIN-76A diet alone or premixed with piroxicam (60 ppm) for 9 weeks. The number, location and volume of tumors, and apoptosis in the flat mucosa were determined in small and large intestine. RESULTS: Piroxicam reduced the number of tumors per mouse by 80% in the small intestine (0.1 vs. 0.5, p < 0.05). In contrast, piroxicam increased tumor incidence (82% vs. 10%, p < 0.01), tumor multiplicity (1.2 vs. 0.1, p < 0.01) and tumor volume (2.1 vs. 0.2 mm3, p < 0.01) in the colon. Apoptosis increased in the epithelium of the small intestine. CONCLUSION: Consistent with the increased apoptosis, piroxicam reduced tumors in the small intestine. In the cecum, piroxicam increased tumorigenesis but apoptosis was not decreased, suggesting that other mechanisms besides apoptosis are involved in the differential organ-specific effect on tumorigenesis of piroxicam in this colon cancer model.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis/drug therapy , Colorectal Neoplasms, Hereditary Nonpolyposis/pathology , Piroxicam/pharmacology , Adaptor Proteins, Signal Transducing/genetics , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/pathology , Genes, APC , Genetic Predisposition to Disease , Intestine, Small/drug effects , Intestine, Small/pathology , Mice , Mice, Mutant Strains , MutL Protein Homolog 1 , Nuclear Proteins/genetics , Random Allocation
12.
Int J Cancer ; 116(4): 495-9, 2005 Sep 10.
Article in English | MEDLINE | ID: mdl-15818612

ABSTRACT

The role of the nuclear peroxisome proliferator-activated receptor-gamma (PPAR-gamma) in colon tumorigenesis remains controversial. Notwithstanding evidence that PPAR-gamma ligands impede murine colorectal carcinogenesis, PPAR-gamma agonists have been shown to enhance in vivo tumor formation in mouse models of human colon cancer. Our study was designed to determine whether troglitazone (TGZ) induces colonic tumor formation in normal C57BL/6J mice and enhances colorectal carcinogenesis in double mutant Apc1638N/+ Mlh1+/- mice fed a standard AIN-76A diet. We report herein that not only does TGZ enhance carcinogenesis in the large intestine of mutant mice predisposed to intestinal carcinogenesis but TGZ also induces colonic tumors in normal mice without gene targeting or carcinogen administration. This observation indicates that preexisting mutational events are not necessary for induction of colonic tumors by activated PPAR-gamma in vivo.


Subject(s)
Antineoplastic Agents/pharmacology , Chromans/pharmacology , Colonic Neoplasms/chemically induced , Thiazolidinediones/pharmacology , Adaptor Proteins, Signal Transducing , Animals , Carcinogens/administration & dosage , Carrier Proteins , Cell Transformation, Neoplastic , Colonic Neoplasms/physiopathology , Colonic Neoplasms/veterinary , Diet , Genes, APC , Mice , Mice, Inbred C57BL , MutL Protein Homolog 1 , Mutation , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , PPAR gamma , Troglitazone
13.
Anticancer Res ; 23(3B): 2217-26, 2003.
Article in English | MEDLINE | ID: mdl-12894495

ABSTRACT

Altered expression of cyclin D1 contributes to the development of several types of cancer, including colorectal cancer. This study examined cyclin D1 expression in 32 intestinal tumors in different stages of tumorigenesis in Apc1638N mice, a mouse model for human familial adenomatous polyposis (FAP). Three morphological patterns of expression of cyclin D1 in intestinal epithelial cells were found: nuclear, punctate-cytoplasmic and fine granular cytoplasmic. The nuclear pattern of cyclin D1 was detected in all of the tumors, including adenomas (n = 18) and adenocarcinomas (n = 14); this pattern was found predominantly in the tubular region of the tumors and in flat mucosa adjacent to a subset of the tumors (67% of adenomas and 57% of carcinomas). The punctate-cytoplasmic pattern of cyclin D1 expression was found in all adenocarcinomas and a majority of adenomas (80%), mainly in invasive and villous areas of the tumors; it was not found in normal flat adjacent mucosa suggesting that this pattern and altered cytoplasmic/nuclear expression were associated with tumor progression. Fine cytoplasmic granules were located in normal duodenum in the basal portion of the crypts and in colon in epithelial cells at the surface of the colonic crypts; in both duodenum and colon the number of cells with fine cytoplasmic granules significantly increased after feeding a Western-style diet. These altered patterns of expression of cyclin D1 may provide useful biomarkers of abnormal cell development for studies of tumorigenesis and the effects of chemopreventive agents.


Subject(s)
Adenomatous Polyposis Coli/metabolism , Cyclin D1/biosynthesis , Intestinal Mucosa/metabolism , Animals , Colon/metabolism , Diet , Duodenum/metabolism , Epithelial Cells/metabolism , Female , Immunohistochemistry , Male , Mice
14.
Carcinogenesis ; 24(3): 605-11, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12663524

ABSTRACT

Sulindac and other NSAIDs have been widely studied as potential chemopreventive agents for colon cancer. Short-term studies have shown adenomatous polyps to regress in patients with familial adenomatous polyposis (FAP). In this study the effect of sulindac on cancer as an endpoint was evaluated in ApcMin mice, a preclinical model of FAP with an Apc mutation in codon 850 that leads to gastrointestinal adenomas and carcinomas. Three groups of mice were studied all of which were fed AIN-76A diet: one group was fed AIN-76A diet alone, a second group received sulindac 200 p.p.m. premixed in the diet and a third group received sulindac 180 p.p.m. added in drinking water. ApcMin mice were killed 9 weeks after feeding was initiated. Mice receiving sulindac developed fewer tumors in the intestine overall; the major decrease in tumor development after sulindac was seen in the small intestine regardless of route of administration. In the large intestine, however, sulindac significantly increased the incidence, multiplicity and volume of tumors in the colon of ApcMin mice, a regional response to sulindac differing from previous reports. Quantitative measurements of apoptosis, Bax and Bcl-xL protein expression in the ApcMin mice revealed the ratio of Bax/Bcl-xL expression and apoptosis increased in the small intestine but decreased in the cecum, consistent with the regional tumorigenesis observed after sulindac. These findings thus suggest involvement of Bax and apoptosis in tumors developing after sulindac treatment in this mouse model.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Genes, APC , Intestinal Neoplasms/chemically induced , Intestine, Large/pathology , Intestine, Small/pathology , Mutation , Proto-Oncogene Proteins c-bcl-2 , Sulindac/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Intestinal Neoplasms/pathology , Intestine, Large/metabolism , Intestine, Small/metabolism , Mice , Proto-Oncogene Proteins/metabolism , Sulindac/adverse effects , bcl-2-Associated X Protein
15.
Proc Natl Acad Sci U S A ; 99(23): 14937-42, 2002 Nov 12.
Article in English | MEDLINE | ID: mdl-12417741

ABSTRACT

Mbd4 (methyl-CpG binding domain 4) is a novel mammalian repair enzyme that has been implicated biochemically in the repair of mismatched G-T residues at methylated CpG sites. In addition, the human protein has been shown to interact with the DNA mismatch repair protein MLH1. To clarify the role of Mbd4 in DNA repair in vivo and to examine the impact of Mbd4 inactivation on gastrointestinal (GI) tumorigenesis, we introduced a null mutation into the murine Mbd4 gene by gene targeting. Heterozygous and homozygous Mbd4 mutant mice develop normally and do not show increased cancer susceptibility or reduced survival. Although Mbd4 inactivation did not increase microsatellite instability (MSI) in the mouse genome, it did result in a 2- to 3-fold increase in C-->T transition mutations at CpG sequences in splenocytes and epithelial cells of the small intestinal mucosa. The combination of Mbd4 deficiency with a germ line mutation in the adenomatous polyposis coli (Apc) gene increased the tumor number in the GI tract and accelerated tumor progression. The change in the GI cancer phenotype was associated with an increase in somatic C-->T mutations at CpG sites within the coding region of the wild-type Apc allele. These studies indicate that, although inactivation of Mbd4 does not by itself cause cancer predisposition in mice, it can alter the mutation spectrum in cancer cells and modify the cancer predisposition phenotype.


Subject(s)
Codon, Terminator/genetics , DNA Repair/genetics , Endodeoxyribonucleases/genetics , Frameshift Mutation , Gastrointestinal Neoplasms/genetics , Animals , Base Pair Mismatch , Base Sequence , Blastocyst/physiology , Chimera , Crosses, Genetic , DNA Primers , DNA Probes , Endodeoxyribonucleases/deficiency , Endodeoxyribonucleases/metabolism , Exons , Female , Gastrointestinal Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Polymerase Chain Reaction , Restriction Mapping , Reverse Transcriptase Polymerase Chain Reaction
16.
Proc Natl Acad Sci U S A ; 99(15): 9924-9, 2002 Jul 23.
Article in English | MEDLINE | ID: mdl-12119409

ABSTRACT

Flap endonuclease (Fen1) is required for DNA replication and repair, and defects in the gene encoding Fen1 cause increased accumulation of mutations and genome rearrangements. Because mutations in some genes involved in these processes cause cancer predisposition, we investigated the possibility that Fen1 may function in tumorigenesis of the gastrointestinal tract. Using gene knockout approaches, we introduced a null mutation into murine Fen1. Mice homozygous for the Fen1 mutation were not obtained, suggesting absence of Fen1 expression leads to embryonic lethality. Most Fen1 heterozygous animals appear normal. However, when combined with a mutation in the adenomatous polyposis coli (Apc) gene, double heterozygous animals have increased numbers of adenocarcinomas and decreased survival. The tumors from these mice show microsatellite instability. Because one copy of the Fen1 gene remained intact in tumors, Fen1 haploinsufficiency appears to lead to rapid progression of cancer.


Subject(s)
Adenocarcinoma/genetics , Endodeoxyribonucleases/deficiency , Endodeoxyribonucleases/genetics , Intestinal Neoplasms/genetics , Mutation , Adenocarcinoma/pathology , Alleles , Animals , Base Sequence , Cloning, Molecular , Codon, Terminator/genetics , DNA Primers , DNA Repair , DNA Replication , Disease Progression , Flap Endonucleases , Frameshift Mutation , Genotype , Intestinal Neoplasms/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data
SELECTION OF CITATIONS
SEARCH DETAIL
...