5v. 2. Hunt..- .232... n .. I: it... an: I .2: V2 .031. 9... r. 4 A .3. fié» (1.2! .% £3: ‘3 . ya- 3.: 4n 4WD... a ti“. .m... .. x: $49 LE1 '1 :3":- . .1 a :4 . t: a... ‘ {at :: . 49 (‘ .33.... .T-.\.\ A 112.... z .x Sin; 1: 1 731.- 2.. .. I}! a) nit); . ‘r‘. 33.3%.: i r .. , .2... i.‘ x. I ‘3‘ ‘ l.v | . .Ipvu... . ma. .3? . a hr... ufii: ,mfifim M? grit-iii: ’K :\4.:» .42.: I! ,1 ’- v ) l5 . .Xy. . 5!..)(l.$~1\31\~( . I I. _ 2': .l of|>|\‘60.lna R i. I“§” UBRARY 20°02: Michigan State University This is to certify that the dissertation entitled MODULATION OF INTESTINAL TUMORIGENESIS BY DIETARY CARBOHYDRATE AND PROTEIN SOURCES IN APCM'N MICE presented by Bing Wang has been accepted towards fulfillment of the requirements for the Doctoral degree in Human Nutrition Mfi mg“; Major Professor’s Signature /2//>"/oS' Date Doctoral Dissertation MS U is an Affirmative Action/Equal Opportunity Institution PLACE IN RETURN BOX to remove this checkout from your record. To AVOID FINES return on or before date due. MAY BE RECALLED with earlier due date if requested. DATE DUE DATE DUE DATE DUE 2/05 p:/ClRC/DateDue.indd-p.1 MODULATION OF INTESTINAL TUMORIGENESIS BY DIETARY CARBOHYDRATE AND PROTEIN SOURCES IN APCMIN MICE By Bing Wang A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Department of Food Science and Human Nutrition 2005 ABSTRACT MODULATION OF INTESTINAL TUMORIGENESIS BY DIETARY CARBOHYDRATE AND PROTEIN SOURCES IN APCMIN MICE By Bing Wang Three experiments were conducted to test the influence of dietary carbohydrate and protein sources on intestinal tumorigenesis in APCMin mice. In the first experiment, APCMin mice consuming sucrose (versus cornstarch) had significantly more but smaller adenomas in the proximal third of the small intestine after 10 weeks of dietary treatment. Mice fed diets containing sucrose (versus comstarch) had a greater rate of colonic epithelial cell proliferation, a greater serum glucose concentrations and hepatic IGF-I mRNA expression, and they also tended (P = 0.07) to have higher serum insulin levels. In the second experiment, global gene expression in small intestinal epithelium caused by APC gene mutation and dietary carbohydrate source was assessed using cDNA microarray analysis. Expression of 379 genes was significantly different between APCMin mice and wild-type mice. Among these differentially-expressed genes, 109 were annotated and had expression altered by genotype by more than 50%. Some of these genes were associated with cell growth control and carcinogenesis (APC gene mutation increased expression of Clu, Ccnd2, Ccnbl , Btg4, Anxal and Gsptl, and decreased expression of Camkld, sept2, LatSZ, Dab2 and Morf4ll). Expression of 306 genes was significantly influenced by dietary carbohydrate source. Among these genes, 87 were annotated and had expression altered by carbohydrate source by more than 50%. Several of these genes also were associated with cell growth control and carcinogenesis (sucrose increased expression of Igf2, Pena, Csell, Idb2 and Cameg, and decreased expression of Igfhp3, Tial, Fancg, Bmprla and Cul4b). Sucrose feeding increased intestinal expression of IGF2 and decreased expression of IGFBP3, which may result in elevated IGF signaling in the intestinal epithelium. In the third study, the influence of dietary carbohydrate (sucrose versus cornstarch) and protein (soy versus casein) sources on colonic adenoma development in APCMin mice was assessed. Sulindac (100 mg/ g) was added to all diets to delay small intestinal tumorigenesis in this study. After a longer duration of dietary treatment (i.e. 16 weeks), APCMin mice consuming sucrose (versus cornstarch) had significantly greater incidence of colon adenomas, as well as increased epithelial cell proliferation and reduced apoptosis in colonic crypts. Female mice consuming soy flour had significantly greater mammary gland tumor incidence (20%) compared with those consuming casein-based diets (0%). ACKNOWLEDGMENTS I would like to sincerely thank my major professor, Dr. Leslie Bourquin, for his guidance and constant support throughout the program. I really appreciate his understanding and encouragement at my difficult time to keep me moving forward. I would also like to thank Dr. John LaPreS for allowing me to use his lab and his assistance with the microarray experiments. I would also like to express my gratitude to Dr. Dale Romsos for his precious advice and kindness. Thanks are also extended to Dr. Maurice Bennink for his help and guidance. I would like to thank Dr. Gerd Bobe and Crystal Ybarra for helping me with animal care and experiments. I thank Elizabeth Rondini for her help and friendship for all the years during my program. I would also like to thank Ajith Vengellur and KangAe Lee for their generous help when I was doing microarray studies. I would like to express my appreciation to my dear wife, Ying Qiu, for her love, support and understanding through all the tough times. Finally, to my three kids, Allen, Joshua and Angelina, they make me feel truly blessed. iv TABLE OF CONTENTS LIST OF TABLES .............................................................................. vii LIST OF FIGURES .............................................................................. x INTRODUCTION ................................................................................ 1 CHAPTER 1. REVIEW OF LITERATURE ................................................................... 3 1. Overview of colorectal cancer .................................................... 4 2. Epithelial cell turnover in colonic crypts .......................................... 5 3. Genetics alterations in colon carcinogenesis .................................... 7 4. APCMln mouse model for cancer research ........................................ 9 5. Dietary carbohydrates and risk for colon cancer ............................. 12 6. Soybean proteins and risk for colon cancer and breast cancer ........................................................................ 25 7. Hypotheses and research objectives ............................................. 28 CHAPTER H. INFLUENCE OF DIETARY CARBOHYDRATE SOURCE (SUCROSE VERSUS CORNSTARCH) ON INTESTINAL TUMORIGENESIS IN APCMIN MICE ...................................................... 31 1. Abstract ............................................................................ 32 2. Introduction ........................................................................ 34 3. Materials and methods ............................................................ 36 4. Results .............................................................................. 46 5. Discussion .......................................................................... 59 CHAPTER III. GENE EXPRESSION PROFILING OF INTESTINAL EPITHELIUM IN APCMIN MICE CONSUMING DIETS CONTAINING DIFFERENT CARBOHYDRATE SOURCES 66 m-wa—d Abstract .............................................................................. 67 Introduction ........................................................................ 69 Materials and methods ........................................................... 71 Results .............................................................................. 8 1 Discussion ........................................................................ 104 CHAPTER IV. INFLUENCE OF DIETARY CARBOHYDRATE (SUCROSE VERSUS CORNSTARCH) AND PROTEIN (CASEIN VERSUS SOY FLOUR) SOURCES ON INTESTINAL TUMORIGENESIS IN SULINDAC- TREATED APCMIN MICE .................................................................. 119 1 Abstract ........................................................................... 120 2 Introduction ...................................................................... 122 3 Materials and methods .......................................................... 124 4 Results ............................................................................. 127 5 Discussion ........................................................................ 145 CHAPTER V. OVERALL SUMMARY AND CONCLUSIONS ......................................... 151 APPENDICES ................................................................... , ............ 1 59 REFERENCE ................................................................................... 189 vi Table 1. Table 2. Table 3. Table 4. Table 5. Table 6. Table 7 Table 8. Table 9. LIST OF TABLES Composition of diets used in experiment 1 (g/kg diet) List of primers used for real time PCR for gene .37 expression in experiment 1 .......................................................... 43 Adenoma incidence, average numbers of adenomas per mouse, total adenoma burden per mouse and avera .e size of adenomas in the small intestine of APC '" mice consuming diets based on differing carbohydrate sources in experiment 1 (means i standard errors) ........................................................... Adenoma incidence, average numbers of adenomas per mouse, total adenoma burden per mouse and average size of adenomas in the colon of APCMm mice consuming diets based on differing carbohydrate sources in experiment 1 (means i standard errors) .................. Ki67 antigen expression in colon of APCMin mice consuming diets containing different carbohydrate sources (means i standard errors) ..................................... PCNA antigen expression in colon of APCMin mice consuming diets containing different carbohydrate sources (means 3: standard errors) ..................................... TUNEL assay results in colon of APCMin mice consuming diets containing different carbohydrate sources (means :1: standard errors) ..................................... Relative expression of IGF-H, IGFBPl and IGFBP3 mRNA in liver of mice consuming diets containing different carbohydrate sources (means 3: standard errors) ..................................................................... List of primers used for confirmatory real time PCR analyses of gene expression ............................................ vii ............. 51 ............. 51 ............. 52 ............. 53 ............. 53 ............. 58 ............. 76 Table 10. Table 1 1. Table 12. Table 13. Table 14. Table 15. Table 16. Table 17. Table 18. Table 19. Table 20. Genes-up-regulated in small intestinal epithelium of M . . . APC m mice versus Wild-type mice ................................... Genes down-regulated in small intestinal epithelium of APCM‘" mice versus wild-type mice Genes up-regulated in small intestinal epithelial cells of mice consuming diets based on sucrose versus cornstarch ................................................................. Genes down-regulated in small intestinal epithelial cells of mice consuming diets based on sucrose versus cornstarch ................................................................. Genes whose expressions were altered differently by ............. 84 .87 ............. 94 ............. 97 APC genotype and dietary sucrose .................................................. 99 Influence of APC gene status on expression of selected genes in small intestinal epithelial cells measured by RT-PCR .............................................................. 102 Influence of APC gene status on expression of selected genes in colon epithelial cells measured by RT-PCR ............................................................................... 102 Influence of dietary sucrose versus cornstarch on expression of selected genes in small intestinal epithelial cells measured by RT-PCR ............................................. 103 Influence of dietary sucrose versus cornstarch on expression of selected genes in colon epithelial cells measured by RT-PCR ................................................................. 103 Composition of diets for experiment used in experiment 3 .......................................................................... 125 Small intestinal adenoma numbers, average sizes of small intestinal adenomas, and total burden of small intestinal adenomas in APCMin mice consuming diets based on differing protein and carbohydrate sources ........................... 131 viii Table 21. Colonic adenoma numbers, average sizes of colonic adenomas, and total burden of colonic adenomas in APCMin mice consuming diets with protein and carbohydrate sources ............................................................... 138 Table 22. Mammary tumor incidence in female APCMin mice consuming diets based on differing protein and carbohydrate sources ................................................................ 139 Table 23. Mean heights of colonic crypts and Ki-67 antigen expression in colon of APCMin mice consuming diets based on differing protein and carbohydrate sources ........................... 142 Table 24. PCNA expression in colon of APCMin mice consuming diets based on differing protein and carbohydrate sources ................................................................ 143 Table 25. TUNEL assay results in colon of APCMin mice consuming diets based on differing protein and carbohydrate sources ................................................................ 144 Table A1. Genes up-regulated in small intestinal epithelium of mice consuming diets based on sucrose versus cornstarch ............................................................................. 160 Table A2. Genes down-regulated in small intestinal epithelium of mice consuming diets based on sucrose versus cornstarch ............................................................................ 167 Table A3. Genesup-regulated in small intestinal epithelium of APCM'" mice versus wild-type mice ............................................... 172 Table A4. Genes down-regulated in small intestinal epithelium of APCM'" mice versus wild-type mice ........................................... 177 Table A5. Genes whose expressions were altered differently by APC genotype and dietary sucrose ................................................ 186 ix Figure 1. Figure 2. Figure 3. Figure 4. Figure 5. Figure 6. Figure 7. Figure 8. Figure 9. Figure 10. Figure 11. LIST OF FIGURES Genetic alterations observed in colon carcinogenesis. Function of APC protein in WNT signaling ........... Proposed mechanism whereby dietary high sucrose increased IGF signaling .................................... IGF signaling pathway and increased risk for colon cancer ......................................................... Weekly body weights of APCMin mice consuming diets containing sucrose or cornstarch as the primary carbohydrate source ........................................ Numbers of small intestinal adenomas in different small intestinal regions of APCM‘" mice consuming diets containing sucrose or cornstarch as the primary carbohydrate source ........................................ Average sizes of small intestinal adenomas (mmz) in different small intestinal regions of APCM'" mice consuming diets containing sucrose or cornstarch as the primary carbohydrate source ......................... Total burden of small intestinal adenomas.(mm2) in different small intestinal regions of APCM'n mice consuming diets containing sucrose or corn starch as the primary carbohydrate source ......................... Glucose concentrations in blood serum of mice consuming diets containing different dietary carbohydrate sources ....................................... Insulin concentrations in blood serum of mice consuming diets containing different dietary carbohydrate sources ....................................... Relative expression of IGF-I mRNA in liver of mice consuming diets containing different dietary carbohydrate sources ...................................... ........................... 8 ......................... 11 ......................... 18 ......................... 22 ......................... 47 ......................... 48 ......................... 49 ......................... 50 ......................... 55 ......................... 56 ........................ 57 Figure 12. Complete loop design for the microarray study ................................. 74 Figure 13 Body weights of APCMin mice consuming diets containing different protein and carbohydrate sources ............................................................................... 130 Figure 14. Main effect of carbohydrates on small intestinal adenoma numbers in APCMin mice consuming diets containing different carbohydrate and protein sources .............................................................................. 132 Figure 15. Main effects of carbohydrates on small intestinal adenoma sizes in APCMin mice consuming diets containing different carbohydrate and protein sources ............................................................................... 133 Figure 16. Main effects of carbohydrates on small intestinal adenoma burden in APCMin mice consuming diets containing different carbohydrate and protein sources ............................................................................... 134 Figure 17 Main effect of proteins on adenoma numbers in APCMin mice consuming diets containing different carbohydrate and protein sources ................................................ 135 Figure 18 Main effect of proteins on adenoma sizes in APCMin mice consuming diets containing different carbohydrate and protein sources ................................................ 136 Figure 19. Main effect of proteins on adenoma burden in APCMin mice consuming diets containing different carbohydrate and protein sources ................................................ 137 xi INTRODUCTION Colon cancer is the second most common cause of cancer mortality and claims more than 50,000 lives every year in the United States (ACS, 2004). Dietary factors strongly modify colorectal cancer risk and approximately 66% to 75% of colon cancer may be preventable by adequate diets and physical activity (AICR/WCRF, 1997). Diets rich in refined carbohydrates such as sucrose have been associated with high colon cancer risk (Tuyns et al. 1988; La Vecchia et al. 1993; Bostick et al. 1994; Centonze et al. 1994; De Stefani et al. 1998). Experiments using carcinogen-induced colon cancer also have shown that feeding rats with high-sucrose diets promoted growth of aberrant crypt foci (ACF), a proposed precancerous lesion of the colon, and also Were associated with increased proliferation in colon crypts compared with feeding high-comstarch diets (Kristiansen et al. 1995; Cademi et al. 1993, 1997; Poulsen et al. 2001). Colon adenocarcinomas in carcinogen-treated rats cosuming high-sucrose diets were Significanatly larger and had more invasive potential compared with those in rats consuming high-comstarch diets (Cademi et al. 1994). However, to date high-sucrose diets have not been demonstrated to increase colon cancer incidence in animal studies. Furthermore, the underlying mechanisms whereby high dietary sucrose may increase colon cancer risk are unclear. APCMin (multiple intestinal neoplasia) mice carry a gemline mutation in the adenomatous polyposis coli (APC) gene (Su et al. 1992) which is similar to APC gene mutations observed in humans with FAP (familial adenomatous polyposis). FAP patients develop hundreds of colonic adenomas and are at increased risk of colon cancer (N ishisho et al. 1991). APC gene mutations also are commonly present in sporadic colon cancers (Powell et al. 1992). Kinzler and Vogelstein (1996) have proposed a “gatekeeper” function for the APC gene in colorectal epithelia in which APC is responsible for maintaining a constant census in renewing cell populations. Therefore, APCMin mice represent a useful model to study diet and gene interactions during colon carcinogenesis. Animal studies suggest that high intakes of sucrose induce compensatory hyperinsulinemia by causing a decline in insulin sensitivity in the liver and peripheral tissues. Hyperinsulinemia may increase IGF-I (insulin-like growth factor 1) production by the liver and thereby increase IGF-l concentrations in the circulation (Giovannucci 2001). Evidence also indicates possible associations between altered gene expressions of IGFBP3 (insulin-like grth factor binding protein 3) and IGF-II (insulin-like growth factor II) in the colon epithelium and colon carcinogenesis (Hassan et al. 2000; Kirman et al. 2004). The purpose of the current research is to determine if diets high in sucrose (vs. cornstarch) promote colon carcinogenesis in APCMin mice, and if this action is associated with elevated IGF signaling and increased cell proliferation in colonic epithelial cells. A secondary objective of this research is to determine the influence of dietary protein source (soy versus casein) on intestinal tumorigenesis in APCMm mice. CHAPTER 1. REVIEW OF LITERATURE 1. Overview of Colorectal Cancer Worldwide, colorectal cancer (CC) is the third most common cancer in women and fourth most common cancer in men. Colorectal cancer incidence varies more than ten-fold across different regions on earth. The highest incidence rates are found in developed regions such as North America, Western Europe, Japan and Australia. In contrast, much lower colon cancer incidences are typically observed in less-developed areas such as Asia and Africa (lARC, 2002). The American Cancer Society estimates that 106,370 new cases of colon cancer and 40,570 new cases of rectal cancer were diagnosed in the United States in 2004. Colorectal cancer is the second most common cause of cancer death in the US, and was expected to cause about 56,730 deaths in 2004, accounting for about 10% of cancer deaths (ACS, 2004). Cancer as a disease is multi-factorial in its causality. Many factors such as hereditary defects, diet, smoking, and exposure to environmental carcinogenic chemicals all may contribute to the generation of cancers. It is estimated that the lifetime risk of developing CC is approximately 5% (Jemal et al. 2002). Most cases of CC arise sporadically (no background of a family history of the disease), but inherited cancer syndromes may account for up to 5% of all CC cases (Ilyas et al. 1999). In 1969, Burkitt first suggested that a low intake of dietary fiber might be involved in the causation of colorectal cancer (Burkitt, 1969). Since that time, abundant epidemiological evidence has revealed that dietary factors strongly influence colorectal cancer incidence. High risk for developing colon cancer has been associated with higher meat and fat consumptions (Stemmerman et al. 1984; Enstrom, 1975), whereas lower risk is associated with consumption of diets rich in vegetables and fruits (Howe ct al. 1992). Dietary composition may be one of the most important factors contributing to cross- cultural differences in colorectal cancer incidence. The American Institute for Cancer Research estimated that about 66% to 75% of colorectal cancer incidence may be associated with inadequate diets and low physical activity (AlCR/WCRF, 1997). 2. Epithelial Cell Turnover in Colonic Crypts The colon can be functionally divided through the transverse colon into two parts, the right and left colon. The right colon (cecum and ascending colon) plays a major role in water and electrolyte absorption and fermentation of undigested sugars, and the left colon (descending colon, sigmoid colon and rectum) is predominantly involved in storage and evacuation of stool. The colon is a muscular organ, and its wall consists of four basic layers — the mucosa, submucosa, circular muscle and longitudinal muscle. The colon mucosa is lined by a simple columnar epithelium that is folded into a number of deep cavities or crypts. The colonic epithelium is in a constant state of cell renewal. A stem cell population located in the base of the crypt proliferates, giving rise to daughter cells. These daughter cells migrate towards the surface of the crypt, and along the way undergo further cell divisions. This cell proliferation is typically localized to the basal two-thirds of normal colonic crypts, which is referred as the proliferation compartment (Lipkin et al. 1974). As the cells migrate toward the crypt surface, they undergo terminal differentiation into a number of cell types, including absorptive cells, mucin-producing goblet cells and endocrine cells. These differentiated cells will continue to migrate toward the luminal surface of the crypt, and ultimately are removed by programmed cell death (apoptosis) at the crypt surface. Apoptosis also may occur to remove cells along the length of the crypt. Colon carcinogenesis is associated with abnormal cell population renewal in the colonic crypts. Deschner (1974) reported that mice injected with the colon carcinogen 1,2-dimethylhydrazine (DMH) had a significantly higher proportion of crypt epithelial cells in S-phase. Furthermore, they noted an expansion of the proliferative compartment towards the luminal surface. Human patients with colon adenomas or cancer also demonstrate abnormal patterns of cell proliferation, with increased proliferation and an upward shifi in the major zone of DNA synthesis (Deschner et al. 1981). Collectively, human studies indicate that increased labeling index (LI) of proliferating cells and an upward shift of the proliferative compartment zone in colonic crypts are biomarkers for increased colorectal cancer risk (Lipkin et al. 1974, 1984; Bleiberg et al. 1985; Terpstra et al. 1987; Ponz de Leon et al. 1988; Mills et al. 2001). However, increased cell proliferation alone has not been directly associated with increased cancer risk (Faber 1995). For example, tumors in the small intestine are extremely rare despite the higher rates of epithelial cell proliferation observed in the small intestine compared to the colon (Faber, 1995). Other processes, such as epithelial cell differentiation and programmed cell death, also have been found to be associated with risk of colonic carcinogenesis (Wyllie, 1985; Boland, 1992). Homeostasis of the colorectal epithelium is dependent not only on the rate of epithelial cell production but also on the rate of apoptosis (epithelial cell removal). Studies have demonstrated that the transformation of colorectal epithelium to carcinomas is associated with a progressive inhibition of apoptosis. The reduced rates of apoptosis observed in colorectal cancers may contribute to tumor growth, promote neoplastic progression, and confer resistance to anticancer agents (Bedi et al. 1995). Thus, colonic neoplasia can be considered as a disease characterized by an abnormal increase of cell population in colonic crypts resulting from disordered regulation of cell proliferation, differentiation and apoptosis. 3. Genetic Alterations in Colon Carcinogenesis Cancer development is a multi-step process consisting of at least three stages — initiation, promotion and progression (F oulds, 195 8). Colorectal cancer development can be categorized by several histological steps, which are described as the "adenoma to carcinoma sequence” (Muto et al. 1975). In this sequence, the smallest lesion is an aberrant crypt focus (ACF), which can be either hyperplastic or dysplastic (N ucci et al. 1997). The most common type of ACF is the hyperplastic crypt, which has increased cell numbers but relatively normal crypt structure and seldom develops into malignant carcinomas. Dysplastic ACF, also termed unicryme adenomas, have abnormal crypt structure and can further develop towards malignancy. Expansion of dysplastic ACFs gives rise to larger adenomas of several centimeters. Adenomas can further progress into carcinoma in situ. At this latter stage, carcinomas eventually may invade adjacent tissues or metastasize to other organs. Fearon and Vogelstein (1992) proposed a genetic model for colorectal tumorigenesis related to their histological stages in the adenoma to carcinoma sequence (Figure 1). Colonic tumorigenesis proceeds through a series of genetic alterations involving mutation of proto-oncogenes such as the ras gene, and inactivation of tumor suppressor genes such as the APC, p53, and DCC (Deleted in Colon Cancer) genes. The APC gene has been proposed as the “gatekeeper” gene for colon carcinogenesis (Volgestein et al. 1996). The majority of dysplastic ACFs bear APC mutations, whereas nonmalignant hyperplastic ACFS often harbor mutations in K-ras (Jen et al. 1994; Nucci et al. 1997). K-ras mutations also commonly occur in small adenomas, which may then become larger and more dysplastic tumors through clonal expansion. Loss of the DCC gene typically occurs at later stages of colorectal tumorigenesis. Mutation of the p53 gene often occurs before metastasis. In addition to these common genetic events, there are likely additional unidentified gene mutations associated with colon carcinogenesis. Each of these mutations confers the affected cell with some growth advantage, allowing it to outgrow other neoplastic cells within the tumor and to become the predominant cell type constituting the neoplasm (clonal expansion). Figure 1. Genetic alterations observed in colon carcinogenesis. Modified from Kinzler KW, Vogelstein B. (1996) Cell 87:159-70 12p qu 17p Other Activation 1055 loss genetic k-ras DCC p53 events Amn‘H mm]: ArlefI‘SmH (mm Metastasis 4. APCMill Mouse Model for Cancer Research (1) Colonrectal cancer: The APCMin (multiple intestinal neoplasia) mouse model has been used extensively for colorectal cancer research in recent years. Min is a gemline mutation in the murine APC (adenomatous polyposis coli) gene (Su et al. 1992), which contains a mutation (Leu to stop) at codon 850 resulting in premature truncation of the polypeptide. This mutation is similar to the APC gene mutations commonly observed in humans with familial adenomatous polyposis (FAP). FAP patients develop hundreds of colonic adenomas and have a very high risk of developing colon cancer (N ishisho et al. 1991). APC gene mutations also are common in sporadic colon cancers (Powell et al. 1992). Kinzler and Vogelstein (1996) have proposed a “gatekeeper” function for the APC gene in colorectal epithelia in which APC is believed to be responsible for maintaining a constant census in renewing cell populations. Therefore, APCMin mice offer an excellent model to study the genetic events leading to colon carcinogenesis following APC gene mutation. Recent studies have revealed several details on how APC mutations lead to colonic tumorigenesis. The APC gene encodes a protein having 2843 amino acids and containing several important functional domains. The middle third of the APC protein contains two classes of B—catenin binding repeats. Through interactions with B-catenin, the APC protein participates in two cellular processes related to colon tumorigenesis (Su et al. 1993). The first process is related to cellular adhesion. Interaction between B- catenin and E-cadeherin is necessary for E-cadherin-mediated cell adhesion between epithelial cells (Kemler, 1993). Disruption of epithelial cell adhesion junctions may contribute to epithelial carcinogenesis. The APC protein also impacts colonic tumorigenesis through its participation in the Wnt signaling pathway. Normally, APC forms an intracellular complex with B-catenin, GSK3B (glycogen synthase kinase-3-l3), and conductin/axin. In the absence of Wnt signaling, B-catenin is phosphorylated by GSK3 [3. Subsequently, it is recognized and degraded through a ubiquitin-proteosome mediated process. Following activation of the Wnt signaling pathway, GSK3B is inactivated and nonphosphorylated B-catenin is released from the complex and accumulates in the cytoplasm. Mutations in the APC gene result in production of truncated APC proteins which lack their domains for B-catenin binding. B-catenin cannot form a complex with truncated APC, GSK3B and conductin/axin, and B—catenin will not be phosphorylated by GSK3B and degraded. Thus, APC mutation causes accumulation of B—catenin in the cytosol. B—catenin proteins in the cytosol form complexes with transcription factors such as T-cell factor (ch) and lymphoid enhancer factor (Let) (Behrens et al. 1996), which translocate into the nucleus and can increase the transcription of many genes having ch binding sites (Figure 2). A comprehensive listing of these target genes is maintained at http://www.stanford.edu/~musse/pathways/targets.html. Some of these target genes play important roles in colon carcinogenesis. These include cyclin D1 (Zhang et al. 1997), matrilysin (Crawford et al. 1999), two components of the AP-l transcription complex — c-jun and fra-l (Mann et al. 1999), c-myc (He 1998) and peroxisome proliferator activated receptor 5 (He et al. 1999). 10 Figure 2. Function of APC protein in WNT signaling. / Intracellular space \ / Intracellular space \ Ubiquitin- mediated roteol sis . y B-catenin TCF Targets I [owe | B-catenin Cyclin 01 \ Nucleus / Normal APC Mutated APC (2) Mammary cancer: Female APCMin mice having the 57BL/6J background occasionally develop mammary gland tumors (Moser et al. 1992, 1993), which indicates a tumor suppressor role for APC within the mammary epithelium. However, the mammary gland tumor incidence observed in APCMin mice is generally low because APCMi’n mice die around 15-16 weeks of age due to rapid small intestinal tumorigenesis. The incidence of spontaneous mammary gland tumors has been reported to be approximately 10% in female mice (Min/+) with a hybrid genetic background (Moser et al. 1992). These mice have a reduced intestinal tumor load and an increased life span when compared to C57BL/6J APCMin mice due to expression of some modifier genes (Moser et al. 1992). ll The APC protein is expressed at high levels within the mammary gland epithelium. Loss of heterozygosity at 5q21 (the chromosomal location of human APC) has been reported in sporadic tumors of the breast (Thompson et al. 1993; Kashiwaba et al. 1994). Reduced APC protein expression also has been demonstrated in human breast cancers (Ho et al. 1999). F uruuchi et al. (2000) reported that somatic APC mutations were present in about 18% of primary breast cancers. J in et al. (2001) demonstrated that the promoter region of the APC gene was frequently hypermethylated in breast cancers, which leads to decreased APC gene expression. Ashkenazi Jews have a high presence of APC 11307K polymorphisms, which renders APC proteins to be genetically unstable and prone to somatic mutation. Ashkenazi Jews have increased risk for colon cancer, and also have high risk of breast cancer when APC Il307K polymorphism is associated with BRCA founder mutations (Woodage et al. 1998). 5. Dietary Carbohydrates and Risk for Colon Cancer (1) Epidemiological and experimental studies: Dietary carbohydrate source and concentration have been previously suggested to influence colon cancer risk. High consumption of refined carbohydrates such as sucrose has been associated with higher colon cancer risk (AICR/WCRF, 1997). Using the Iowa Women’s Health Study cohort, Bostick et al. (1994) reported that consumptions of diet containing non-dairy sucrose were associated with increased colon cancer risk (RR = 2.0 for highest versus lowest quintiles, CI = 121-33). The relative risk of colon cancer risk was 1.74 for highest versus lowest quintiles of total sucrose-based food (CI = 1.06-2.87; Bostick et al. 1994). Several case-control studies have reported significant associations between diets high in 12 sucrose with increased risk of colorectal cancer (Bristol et al. 1985; Tuyns et al. 1988; La Vecchia et al. 1993; Centonze et al. 1994; De Stefani et al. 1998). However, other studies found no relationship or only weak associations between sucrose intake and colorectal cancer with odds ratios between 1.0 and 1.3 (Manousos et al. 1983; Macquart- Moulin et al. 1986; La Vecchia et al. 1988; Peters et al. 1992). Results from experimental studies also suggest that higher sucrose intakes are associated with increased colon cancer risk. Feeding sucrose is associated with greater proliferation in colonic crypts, as well as a greater number of large aberrant crypt foci (Cademi et al. 1993, 1997; Kristiansen et al. 1995; Poulsen et al. 2001). Feeding rats for 4 weeks with dietary sucrose versus cornstarch (46%) significantly increased colonic epithelial cell proliferation (Cademi et al. 1993). Rats treated with the colon carcinogen dimethylhydrazine (DMH) had significantly greater numbers of dysplastic aberrant crypt foci (ACF) in colon when consuming diets containing sucrose (46%) compared to those consuming cornstarch (Cademi et al. 1991, 1997). DMH-injected rats fed high-sucrose diets (61%) had significantly greater numbers of ACF during the post-initiation period compared to rats fed starch diets (Pulsen et al. 2001). Cademi et al. (1994) reported that rats fed high sucrose diets had significantly greater numbers of colonic adenomas per rat compared with those fed comstarch-based diets in a DMH-induced rat colon cancer model. The adenocarcinomas in the rats fed sucrose were larger and had greater invasive potential than those in rats fed the comstarch-based diets, although the incidence of total intestinal tumors was not affected by the different carbohydrate sources (Cademi et al 1994). 13 Cademi et al. (1996) studied the influence of different dietary carbohydrate sources on colon lumenal environment. Female rats were fed for one month with diets containing different carbohydrates (sucrose, glucose, fructose, cornstarch, and Hylon 7, a starch with a high amylose content). Colon epithelial cell proliferation was significantly greater in rats fed sucrose than in rats fed glucose, fructose, or cornstarch. Cecal pH was lower in rats fed cornstarch and Hylon 7 than in rats fed sucrose, glucose, or fructose. Cecal concentrations of short-chain fatty acids (SCFAS) were greater in rats fed Hylon 7 than in those fed glucose and fructose (Cademi et al. 1996). In this study, no association was found between the influence of carbohydrate source on colon epithelial cell proliferation and cecal SCF A concentrations or cecal pH (Cademi et al. 1996). Stamp et al. (1993) reported that oral gavages of either sucrose or fructose in mice (10 g/kg body weight) increased colonic epithelial cell proliferation (measured 16 h after gavage) when compared to mice gavaged with an equivalent amount of glucose. Sucrose and fructose, when administered 14 h prior to azoxymethane (AOM) injection, increased average numbers of ACF in colon compared with glucose. The effects of dietary carbohydrates on blood glucose levels vary depending upon the glucose content of the carbohydrate and its rate of digestion, which can be predicted by glycemic index (Jenkins et al. 1981; Wolever et al. 1991). Corpet et al. (1998) studied the influence of diets having different glycemic indices on development of insulin resistance and ACF in rats. After receiving an AOM injection, rats were randomly assigned to AlN-76-based diets containing different carbohydrate sources (65% starch, glucose or fructose). The average numbers of ACF per rat were not different among rats 14 fed starch, glucose or fructose. Indirect markers of insulin resistance did not correlate with ACF multiplicity in this study (Corpet et al. 1998). (2) High consumption of sucrose and hyperinsulinemia: Although sucrose has a lower glycemic index than some complex carbohydrates such as white bread (Jenkins et al. 1981; Wolever et al. 1991), animal studies have found that high intakes of sucrose, relative to cornstarch, cause a decline in insulin sensitivity in the liver and later in peripheral tissues as assessed by euglycemic hyperinsulinemic clamps (Pagliassotti et al. 1995; Storlien et al. 1988; Martinez et al. 1994; PamieS-Andreu et al. 1995). Pagliassotti et al. (1995) reported that male Wistar rats consuming diets containing relatively low levels of sucrose (18% of energy) also developed insulin resistance after 8 weeks when compared to rats consuming cornstarch. The decreased insulin sensitivity caused by high sucrose diets is likely related to the fructose component of sucrose (Thorbum et al. 1989; Thresher et al. 2000). High fructose intakes promote the development of hypertriglyceridemia (Zavaroni et al. 1982; Thorbum et al. 1989). Rats fed high-fructose diets (35%), compared to rats fed glucose, had increased fasting triglyceride levels within 2 weeks after initiation of dietary treatments, and increased triglyceride levels in circulation may be responsible for the development of impaired insulin actin in liver and peripheral tissues (Thorbum et al. 1989). High-fructose diets stimulated hepatic VLDL-TG secretion due to the increased formation of glycerol-3-phosphate, a precursor of lipid synthesis (Zavaroni et al. 1982). Increased plasma concentrations of nonesterified fatty acids may reduce insulin sensitivity by increasing the intramyocellular lipid content (Virkamaki et al. 2001). Increased portal delivery of nonesterified fatty acids increases hepatic glucose production 15 (Rebrin et al. 1995; Steil et al. 1998). High intakes of fructose also cause decreased production of the adipocyte protein, adiponectin. Decreased circulating concentrations of this hormone are associated with insulin resistance independently of body adiposity (Weyer et al. 2001; Havel et al. 2002). However, human studies examining the ability of dietary sucrose to induce insulin resistance have not been convincing, possibly due to limited variation in dietary sucrose intakes, heterogeneity in study populations and complexities of study design (Giovannucci, 2001). Some studies showed that high intakes of sucrose or fructose increased fasting or postprandial insulin concentrations (Reiser et al. 1979, 1981). Other studies indicated that high fructose intakes (20% of carbohydrate, 45-65 g/day for 4 weeks) increased insulin sensitivity (Koivisto et al. 1993), whereas still others showed no effect (Dunnigan et al. 1970). Bessesen (2001) suggested that most of these human studies were conducted using patients who already had insulin resistance, and the effects of diets containing high sucrose or fructose concentrations would more likely be detected in younger individuals who do not already have some degree of insulin resistance. (3) Elevated IGF signaling and increased colon cancer risk: McKeown-Eyssen (1994) and Giovannucci (1995) noted the similarity in geographic distribution of diabetes and colon cancer patients. Patients having either diabetes or colon cancer often share common risk factors such as high body mass index, increased central obesity, physical inactivity, excessive intakes of energy and dietary patterns that increase risk of hyperinsulinemia. Epidemiological studies also found that subjects diagnosed with type II diabetes have increased risk of colon cancer (La Vecchia et al. 1997; Le Marchand et 16 al. 1997; Will et al. 1998; Hu et al. 1999). Based on these observations, it has been proposed that hyperinsulinemia promotes colorectal carcinogenesis. Increased insulin levels were originally thought to promote colon cancer development. However, insulin has been found to be a relatively weak mitogen in vitro, acting as such only at very high concentrations (Giovannucci 1995). Alternatively, Giovannucci (1995, 2001) proposed that the role of insulin in colorectal cancer is mediated through IGF-I. Insulin has been found to increase bioactive IGF-I through various mechanisms (Figure 3). Growth hormone up-regulates the production of IGF-I in liver, and insulin increases IGF-I expression by up-regulating hepatic growth hormone receptor number (Jones et al. 1995; Underwood et al. 1994). Insulin also reduces hepatic secretion of IGFBPl (insulin-like growth factor binding protein 1; Ooi et al. 1992), which in turn will increase the availability of IGF-I to the receptors of other tissues. The IGF signaling system includes two insulin-like growth factors (IGF-I and IGF ~11), which exert their actions by interacting with specific receptors for IGF-I and IGF-II on the cell membrane, and their actions are regulated by a group of specific binding proteins (i.e., IGFBP] through IGFBP6). In addition, a large group of IGFBP proteases hydrolyze IGF BPS, resulting in the release of bound IGFs that then resume their ability to interact with receptors (Stewart et al. 1996; Rajaram et al. 1997). 17 Figure 3. Proposed mechanism whereby high dietary sucrose increases IGF signaling. Modified from Giovannucci (2001) J. Nutr. 131:3 109S-3120S. High sucrose diet Growth hormone Hepatic growth Insulin P hormone ——> _ receptor V V IGFBPl ................................................................... , IGF-I *Solid lines indicate stimulating effect and broken lines indicate inhibition. IGF-I is a potent mitogen and promotes growth of a wide variety of cells. IGF-I also inhibits cell differentiation and apoptosis (Jones et al. 1995; Yu et al. 2000). Like IGF-I, IGF-II has similar effects on cell proliferation, differentiation and apoptosis, and this effect is also achieved through activation of the IGF-IR (Yu et a1. 2000). In contrast, the IGF-IIR has no tyrosine kinase activity, and its binding with IGF-II results in degradation of IGF-II, thereby decreasing IGF-II activity (Stewart et al. 1996; Rajaram et al. 1997). IGF-I and IGF-II in the circulation are primarily produced in liver. Although IGF-II plays a key regulatory role during embryonic and fetal growth, IGF-II plays a less 18 important role in postnatal growth compared with IGF-I (Jones et al. 1995). Levels of circulating IGF-I are mainly regulated by growth hormone and change substantially with developmental stages. IGF-I expression also is altered by nutritional state. For instance, fasting reduces serum IGF-I concentrations as much as 75% and negates the effects of growth hormone (GH) stimulation. Refeeding rapidly restores IGF-I levels to normal (Clemmons et al. 1981). Circulating levels of IGF-II are relatively stable after puberty, and GH has little influence on them (Jones et al. 1995). Actions of IGF-I and IGF-II on growth both are mediated through binding with IGF-IR. However, IGF-IR binding affinity with IGF-I is about 2-15 fold higher than that with IGF-II (Jones et al. 1995). Three of the six IGF BPS have higher affinity to IGF-II than to IGF-I, and the rest have similar binding affinity to both IGFs (Rajaram et al. 1997). The combination of high affinity to the receptor and low affinity to the binding proteins results in more IGF-I than IGF-II interacting with IGF-IR in tissues. Although it is believed that circulatory IGF-I, mainly produced in liver, is most important for postnatal growth, research also has identified the importance of extrahepatic production of IGF-I in growth regulation. Yakar et al. (1999) used the Cre/loxP recombination system to Specifically delete the liver IGF-I gene one to two weeks after birth, and reduced circulating IGF-I concentrations by approximately 80% in mice. The growth rates of these transgenic animals were not significantly different when compared with the wild-type animals. This provided direct evidence for the importance of the autocrine/paracrine role of IGF-I in the extrahepatic tissues. The IGF-II gene is known to have parental allele-specific expression. In normal cells, IGF-II is maternally imprinted in that it is expressed only from the paternal copy of 19 the gene. Loss of IGF-II imprinting has been reported in a variety of tumors (Yu et al. 2000). When loss of imprinting occurs, biallelic expression of IGF-II results in over- expression of this potent growth factor. Activation of the IGF signaling pathway greatly impacts cell proliferation, differentiation, and apoptosis (Jones et al. 1995). Both IGF-I and IGF-II can activate the IGF-I receptor, leading to receptor autophosphorylation and phosphorylation of downstream proteins such as insulin receptor substrate (IRS). IRS serves as docking sites for other cellular proteins and subsequently activates downstream signal transduction pathways (Yu et al. 2000). At least two distinct signal transduction pathways have been identified for IGF-IR: one is the MAPK (mitogen-activated protein kinase) pathway, and increased MAPK pathway signaling leads to increased cell proliferation. Another is the PI3K pathway, wherein activation of Akt results in the phosphorylation of several other proteins that affect cell growth and cell survival. Phosphorylation of the F orkhead transcription factor (FOXO) blocks transcription of P27kip, and decreased expression of P27kip contributes to increased cell proliferation. Phosphorylation of the apoptosis- inducing protein Bad prevents cells from undergoing apoptosis. A third target of Akt is glycogen synthase kinase 3 (GSK3). Phosphorylation of GSK3 (both alpha and beta isoforms) by Akt turns off its catalytic activity. GSK3B is required for phosphorylation and degradation of B-catenin in the Wnt signaling pathway. Inactivation of GSK3B by Akt also leads to the accumulation of B-catenin, which leads to increased cell proliferation (Foulstone et al. 2005; Figure 4). Recent research has indicated that various components in the IGF system are involved in colon cancer development. Using in vitro studies, IGF-I and IGF-II have 20 been demonstrated to stimulate growth of various colon cancer cell lines (Koenuma et al. 1989; Lahm et al. 1992). IGFBP3 is found not only to regulate the mitogenic action of IGFS but also to inhibit their antiapoptotic effect (Stewart et al. 1996; Rajaram et al. 1997) Studies examined circulating IGF-I and IGFBP3 levels in relation to colon cancer risk in humans and found that high blood IGF-I and low IGFBP3 levels are associated with increased colon cancer risk (Ma et al. 1999; Giovannucci et al. 2000). Studies also have demonstrated a direct role of IGFS in colon cancer development. Wu et al. (2002) demonstrated that circulating IGF-I levels regulate colon cancer growth and metastasis in a mouse model of colon cancer. 21 Figure 4. IGF signaling pathway and increased risk for colon cancer. .\/ IGFBPS / l Extracellular Space Q% L n IGFBP proteasefl llllllllllllllllllIlllll llllll ll llll llll Illl lll311lllllllllllllllll Pll-4,5-P2 Pll-3,4,5-P3 Intracellular /‘\ space @ Grb2 @éypnu fl; K @ MAPK p27kiP GSK3B L - WNT Pathway l . . . M / I proliferation j proliferation I Apoptosrs Proliferation / Some studies also have reported associations between local expressions of components of the IGF signaling system in colonic epithelium with colon carcinogenesis. Both IGF-I and IGF-II mRNA levels in colon tumors were found to be slightly elevated compared with normal colonic mucousa (Tricoli et al. 1986). Other studies did not observe significant differences in IGF-IR and IGF-11R expression when comparing colon tumors and normal epithelial cells (Adenis et al. 1995; Zenilman et al. 1997). However, IGF-IR overexpression has been reported in colon cancers, and its overexpression was associated with aggressive tumors (Hakam et al. 1999). In ApcMin mice, it was found that 22 Mm“ mice with IGF-II gene reducing IGF-II expression in the intestine by crossing Ape knockout mice resulted in reduced adenoma size and frequency. On the other hand, inducing over-expression of IGF-II in the intestine significantly increased tumorigenesis in ApcMW+ mice (Hassan et al. 2000). IGFBP3 has been demonstrated to inhibit cancer cell growth induced by IGF-I by sequestering IGF-I from binding IGFI-R, but IGFBP3 also has functions independently from binding IGF-I (Cubbage et al. 1990). A mutated form of IGFBP3 that is unable to bind IGF-I or IGF-II still inhibited tumor growth (I-Iong et al. 2002). In addition, IGFBP3 induces tumor cell apoptosis even in the absence of IGF-I receptors (Valentinis et al. 1995). Recently, Kirman et al (2004) further demonstrated that IGFBP3 inhibits the development of colonic tumors in experimental models. In the study, two tumor models were used. In AOM-induced colon carcinogenesis, wild-type and IGFBP3 transgenic mice (with over-expression of IGFBP3 in tissues) were injected with AOM and the numbers of aberrant crypt foci (ACF) in the colon of transgenic mice were significantly lower compared with that in the wild-type mice. In the syngeneic model, mice were inoculated with CT26 colon cancer cells. The control group received saline, while the test group was injected with IGFBP3 around the tumors weekly. CI26 tumors were significantly smaller in mice that received IGFBP3 (Kirman et al. 2004). Studies have examined circulating IGF-I and IGFBP3 levels in relation to colon cancer risk in humans and found that high blood IGF-I and low IGFBP3 levels are associated with increased colon cancer risk (Ma et al. 1999; Giovannucci et al. 2000). Studies also have demonstrated a direct role of IGFS in colon cancer development. Wu et 23 al. (2002) demonstrated that circulating IGF-I levels regulate colon cancer growth and metastasis in a mouse model of colon cancer. Some studies also have reported associations between local expression of components of the IGF signaling system in colonic epithelium with colon carcinogenesis. Both IGF-I and IGF-II mRNA levels in colon tumors were found to be slightly elevated compared with normal colonic mucousa (Tricoli et al. 1986). Other studies did not observe significant differences in IGF-IR and IGF-IIR expression when comparing colon tumors and normal epithelial cells (Adenis et al. 1995; Zenilman et al. 1997). However, IGF-IR overexpression has been reported in colon cancers, and its overexpression was Min associated with aggressive tumors (Hakam et al. 1999). In Apc mice, it was found that Min/I mice with IGF-II gene reducing IGF-II expression in the intestine by crossing Apc knockout mice resulted in reduced adenoma size and frequency. On the other hand, inducing over-expression of IGF-II in the intestine significantly increased tumorigenesis in ApcMim mice (Hassan et al. 2000). IGFBP3 has been demonstrated to inhibit cancer cell growth induced by IGF-I by sequestering IGF-I from binding IGFI-R, but IGFBP3 also has functions independently from binding IGF-I (Cubbage et al. 1990). A mutated form of IGFBP3 that is unable to bind IGF-I or IGF-II still inhibited tumor growth (Hong et al. 2002). In addition, IGFBP3 induces tumor cell apoptosis even in the absence of IGF-I receptors (Valentinis et al. 1995). Recently, Kirman et al (2004) further demonstrated that IGFBP3 inhibits the development of colonic tumors in experimental models. In the study, two tumor models were used. In AOM-induced colon carcinogenesis, wild-type and IGFBP3 transgenic mice (with over-expression of IGFBP3 in tissues) were injected with AOM and the numbers of aberrant crypt foci (ACF) in the 24 colon of transgenic mice were Significantly lower compared with that in the wild-type mice. In the syngeneic model, mice were inoculated with CT26 colon cancer cells. The control group received saline, while the test group was injected with IGFBP3 around the tumors weekly. CT26 tumors were significantly smaller in mice that received IGFBP3 (Kirman et al. 2004). 6. Soybean Proteins and Risk for Colon Cancer and Breast Cancer In the United States, the incidence and mortality of colorectal, breast and prostate cancers are higher than that observed in certain Asian countries such as Japan and China (ACS, 1994). Since persons in those countries consume more soybean products than Americans, consuming large amounts of soybean products has been suggested to be associated with overall low cancer incidence and mortality rates (Messina et al. 1994; Barnes et al. 1995). However, results from several epidemiological studies were not supportive of a protective role of dietary soy against colon cancer. In a case-control study in China, Hu et al. (1991) only found a protective effect of soybean products against rectal cancer risk in man. Another study in Japan also only found that consumption of beans and bean curd was associated with lower rectal cancer risk (Watanabe et al. 1984). However, in a cross-cultural study of 38 countries, no association between soybean intake and colon cancer risk was found (McKeown-Eyssen et al. 1984). Numerous studies have been conducted to examine the effects of specific components in soybeans on colon cancer risk. Several compounds in soy products have been shown to suppress carcinogenesis in vitro and in vivo. These compounds include the 25 isoflavone genistein, the Bowman-Birk protease inhibitor, inositol hexaphosphate (phytic acid), saponins, and phytosterols such as B-sitosterol (Messina et al. 1994; Kennedy et al. 1995) Results of studies examining effects of soy consumption on colon cancer risk using carcinogen-induced colon cancer models are inconsistent. Thiagarajan et al. (1998) reported that feeding defatted soy flour and full-fat soy flakes significantly reduced the numbers of ACF in azoxymethane (AOM)-treated rats. Bennink et al. (1999, 2000) found that feeding soy flour reduced colon tumor incidence during the promotional phase of carcinogenesis. In another study, Hakkak et al. (2001) reported that the second generation of rats had a significantly lower colon tumor incidence after feeding rats with soy protein isolate for two generations. However, feeding soy protein was not protective against colon carcinogenesis in several other studies using animal models of colon cancer. Feeding soybean meal tended to increase tumor incidence in carcinogen-induced colon cancer in rats compared with feeding casein (P = 0.12; McIntosh et al. 1995). Feeding soy protein isolates containing high levels of isoflavones was found to increase the number of small ACF (less than 4 crypts per foci) after 12 weeks of dietary treatment compared with feeding soy protein having low isoflavone concentrations, although the final colon tumor incidence was not different (Davies et al. 1999). Few human clinical trials with soy products have been conducted to evaluate their potential to protect against colon cancer risk. One double blind prospective study with forty-seven subjects found that dietary supplementation with 39 g per day of isolated soy protein for one year significantly reduced the proliferative capacity in colonic crypts 26 compared with supplementation with an equivalent amount of casein (Thiagarajan et al. 1999) Soy protein consumption also has been associated with reduced risk of breast cancer. It has been reported that modest soy consumption was associated with a 50% reduction in premenopausal breast cancer risk (Lee et al. 1991). However, most epidemiological studies in postmenopausal women found no significant protection of soy consumption (Messina et al. 2001). The soybean isoflavone genistein is often considered to be the component in soy that is primarily responsible for its putative protective effects against breast cancer development. Administration of high concentrations of genistein to female rats during neonatal development or during the prepubertal period reduces the later development of DMBA-induced mammary tumors (Lamartiniere et al. 1995; Murrill et al. 1996). The protective effect of early genistein exposure against mammary cancer is due to promotion of earlier mammary gland maturation, thereby rendering these animals less susceptible to mammary carcinogenesis when exposed to the carcinogen later in life (Messina et al. 2001) Feeding soy products has been reported to reduce mammary tumor incidence or tumor multiplicity compared with feeding casein-based diets when given during tumor initiation (Barnes et al. 1990; Hawrylewicz e t al. 1991; Hakkak et al. 2000). Studies also have investigated effects of soy consumption on mammary tumor progression. Hawrylewicz et al. (1995) reported that, after inducing breast tumors by injecting the carcinogen MN U, rats fed soy protein isolate developed significantly fewer tumors than rats fed casein. Furthermore, tumors in the soy-fed rats were less aggressive than tumors 27 observed in the casein-fed rats (Hawrylewicz et al. 1995). However, Hsieh et al. (1998) found that dietary genistein (750 ug/g) stimulated the growth of subcutaneously implanted MCF-7 cells in ovariectomized athymic mice. Feeding soy protein containing various amounts ofgenistein (15, 150 and 300 ug/g) also has been demonstrated to increase implanted mammary tumor growth in a dose-dependent manner (Allred et al. 2001) Effects of feeding isoflavones on mammary cancer development also were studied using WTV—neu mice, which spontaneously develop mammary tumors due to overexpression of the ErbB-2/neu/HER2 oncogene (J in etal. 2002). Feeding MMTV-neu mice with diets containing different levels of isoflavones from 7 weeks of age, J in et al (2002) found that mammary tumor latency was significantly delayed in mice fed isoflavones compared with the control. However, once tumors formed, isoflavone feeding did not reduce the number or size of tumors such that at 34 weeks of age there were no differences in tumor burden among the treatment groups (J in et al. 2002). 7. Hypotheses and Research Objectives Dietary factors strongly modify colorectal cancer risk and approximately 66% to 75% of colon cancer may be preventable by adequate diets and physical activity (AICR/WCRF 1997). Improving our understanding of the influence of dietary carbohydrates on risk of colon cancer is of great importance. Although epidemiological research and results of animal studies suggest that dietary carbohydrates influence colon cancer risk, the effects of high dietary sucrose have not been tested in APCMin mice. 28 APCMin mice carry a gemline mutation in the APC gene and develop multiple intestinal adenomas that can progress to adenocarcinomas. APC gene mutation is considered as the most critical step in the multi-step colonic carcinogenic process, in which it acts as a “gatekeeper” to ensure cell population census in intestinal crypts (Kinzler and Vogelstein, 1996). Therefore, APCMin mice present a useful model to study diet and gene interactions during colon carcinogenesis following APC mutation. Animal studies suggest that high intakes of sucrose induce compensatory hyperinsulinemia by causing a decline in insulin sensitivity in the liver and peripheral tissues (Pagliassotti et al. 1995; Storlien et al. 1988; Thorbum et al. 1989; Pagliassotti et al. 1994; Martinez et al. 1994; Pamies-Andreu et al. 1995). Hyperinsulinemia can cause changes in the IGF signaling system, such as increased IGF-I expression in liver. Evidence also indicates possible associations between high sucrose intakes and altered gene expressions of IGFBP3 and IGF-II in colon epithelial cells and colon carcinogenesis. The central hypothesis of this research is high dietary sucrose promotes colon carcinogenesis in APCMin mice, and this effect is mediated in part through elevating IGF signaling in the intestinal epithelium. To test this hypothesis, experiments were conducted to 1) test if high dietary sucrose promotes intestinal tumorigenesis in APCMin mice; 2) determine the patterns of cell kinetic alterations in colonic crypts which indicate increased risk of colon cancer; 3) determine if high dietary sucrose changes levels of glucose and insulin in the blood, and mRNA levels of IGF-I, IGF-II and IGFBP] and IGFBP3 in liver; and 4) explore the patterns of global gene expression alterations associated with increased intestinal tumorigenesis in APCMI" mice. Results of these 29 studies will determine if high dietary sucrose increases colon cancer risk in APCMin mice, and also identify potential mechanisms whereby dietary sucrose increases risk for colon cancer. A secondary objective of this research was to examine the effects of dietary protein source on intestinal tumorigenesis in APCMin mice. We hypothesized that dietary soy, compared to casein, reduces intestinal tumorigenesis in APCMin mice. To test this hypothesis, we designed one feeding experiment as a 2 x 2 factorial to test the effects of, and interactions between, dietary carbohydrate (sucrose vs. cornstarch) and protein (soy vs. casein) sources on intestinal tumorigenesis APCMm mice. 30 CHAPTER II. INFLUENCE OF DIETARY CARBOHYDRATE SOURCE (SUCROSE VERSUS CORNSTARCH) ON INTESTINAL TUMORIGENESIS IN APCMIN MICE 31 ABSTRACT Epidemiological evidence suggests that consumption of large amounts of refined carbohydrates may be associated with increased risk of colon cancer. The objectives of this study were to test the effects of diets containing sucrose or cornstarch as the sole carbohydrate source on intestinal adenoma development in APCMin mice, and possible associations between elevated serum IGF concentrations caused by high dietary sucrose and increased colon cancer risk. APCMin mice (n = 48) were randomly assigned at 4 weeks of age to one of two modified AIN-93G diets containing either sucrose or cornstarch (523 g/kg of diet) as the sole carbohydrate source. Diets were fed ad libitum for 10 weeks. Weekly body weights, intestinal adenoma development and epithelial cell kinetics (Ki67, PCNA and TUNEL labeling) in colonic crypts were measured. The levels of glucose and insulin in the circulation, as well as mRNA expressions of IGF-I, IGF-II, IGFBP] and IGFBP3 in liver also were determined. In comparison to cornstarch, sucrose promoted weight gain in mice (P < 0.05). APCMin mice fed sucrose had significantly greater numbers of adenomas in the proximal third of the small intestine when compared to mice fed cornstarch (21.8 vs. 13.1, P < 0.01), however, the average Size of adenomas in the proximal third of the small intestine were larger in cornstarch-fed mice (2.1 vs. 1.1 mmz, P < 0.01). The total number of adenomas in the small intestine tended to be greater in sucrose-fed mice (72 vs. 60, P < 0.07). Colon tumor incidence was 56% in APCMin mice fed sucrose compared with 43% in those fed cornstarch, but this difference was not statistically significant. Mice consuming sucrose had significantly greater Ki-67 antigen labeling index in colonic crypt epithelial cells compared to mice consuming cornstarch (37% vs. 32%, P < 0.05), which 32 indicated that mice consuming sucrose had a greater overall rate of cell proliferation in the colon. Mice consuming the high-sucrose diet had greater serum glucose concentrations than mice consuming cornstarch (10.09 vs. 8.08 mM, P < 0.05). Mice consuming sucrose tended to have higher serum insulin levels compared to mice consuming cornstarch (262.6 vs. 174.4 pmol/L, P = 0.07). Relative IGF-I mRNA expression in liver of mice fed sucrose was greater than that observed in mice fed cornstarch (0.166 vs. 0.092, p = 0.05). Hepatic mRNA expressions of IGF-II, IGFBP] and IGFBP3 were not influenced by diet. In summary, dietary carbohydrate source significantly influenced the numbers and sizes of adenomas in the proximal small intestine. Although carbohydrate source did not influence colon tumor incidence after 10 weeks of treatment, dietary sucrose significantly increased the proliferation rate of colonic epithelial cells. The effects of dietary sucrose on intestinal tumorigenesis may be related to increased levels of IGF-I mRNA expression in liver. 33 INTRODUCTION Colon cancer is the second most common cause of cancer mortality and claims more than 50,000 lives every year in the United States (ACS, 2004). Dietary factors strongly modify colorectal cancer risk and approximately 66% to 75% colon cancer may be prevented by adequate diets and physical activity (AICR/WCRF, 1997). High dietary intakes of refined carbohydrates such as sucrose are associated with higher colon cancer risk (Bostick et al. 1994; De Stefani et al., 1998). Furthermore, recent studies indicate that the there is an increased consumption of simple sugars such as high fructose corn syrup in the United States (Bray et al., 2004). Using US Department of Agriculture food consumption tables from 1967 to 2000, Bray et al. (2004) found that the consumption of high fructose corn syrup increased more than ten-fold between 1970 and 1990, far exceeding the changes in intake of any other food or food group. Experiments using carcinogen-induced colon cancer models have shown that feeding rats with high sucrose diets promoted growth of aberrant crypt foci (ACF), precancerous lesions in colon, and also were associated with increased epithelial cell proliferation in colon crypts compared with feeding cornstarch diets (Kristiansen et al. 1995; Cademi et al. 1993, 1997; Poulsen et al. 2001). Colon adenocarcinomas in carcinogen-treated rats cosuming high-sucrose diets were significanatly larger and had more invasive potential compared with those in rats consuming cornstarch (Cademi et al. 1994). However, to date high sucrose diets have not been demonstrated to increase colon cancer incidence in animal studies. Furthermore, the underlying mechanisms whereby high dietary sucrose may increase colon cancer risk are not clear. Studies have focused on the possible influence of intestinal lumenal environment through fermentation and 34 production of short chain fatty acids (Cardemi et al. 1994). Because sucrose and cornstarch are digested and their constituent monosaccharides are absorbed in the small intestine, any influence on colon carcinogenesis is more likely to be mediated by circulatory factors. APCMin mice carry a mutation in one allele of the APC gene and develop numerous adenomas in the intestine. APC gene mutations are a frequent and early event in colon carcinogenesis. Thus, APCMin mice represent an excellent model to study diet and gene interactions during colon carcinogenesis. The influence of dietary carbohydrates on intestinal tumorigenesis in APCMin mice has not been reported. The first objective of the current study was to determine the influence of dietary carbohydrate source (sucrose vs. cornstarch) on intestinal tumorigenesis in APCMin mice. A second objective was to evaluate serum concentrations of insulin and IGF, as well as hepatic expression of mRNA for IGF-I, IGF-II, IGFBP] and IGFBP3 and their association with intestinal tumorigenesis. Our final objective was to measure the influence of dietary carbohydrate source on cell proliferation and programmed cell death in colonic epithelium. 35 MATERIALS AND METHODS Animals: Due to the high cost and limited commercial availability of APCMin mice, we maintain a breeding colony of APCMin mice at Michigan State University. Mice for the study were produced by mating C57BL/6J male APCMin mice with normal C57BL/6J female mice. The resulting offspring (50% normal and 50% APCMin based on simple Mendelian inheritance) were randomly assigned at weaning (4 weeks of age) to one of two diets, which they were fed for 10 weeks or until they became morbid (> 10% weight loss). Fifty mice were assigned to each treatment. Of these, we expected 50% to be APCMin mice and 50% to have normal APC gene status. Mice used in this study were genotyped by an allele-specific PCR procedure to identify APCMin progeny (Jacoby et al., 1996). Mice were housed in a room with constant temperature and humidity (23°C, 60% humidity) and a 12:12 hour light-dark cycle. Animal care and feeding were conducted with approval of the Michigan State University All-University Committee on Animal Use and Care. Diets: Experimental diets were formulated based on AIN-93G diets (Reeves et al., 1993) and contained either sucrose or cornstarch as the sole carbohydrate source (Table l). Mice were fed diets ad libitum for the duration of the study (ten weeks or until individual animals exhibited body weight loss of more than 10%). 36 Table 1. Composition of diets used in experiment 1 (g/kg diet). Ingredient Corn Starch Diet Sucrose Diet Casein 221 221 Corn Starch 523 0 Sucrose 0 523 Soybean Oil 150 150 Cellulose 50 50 AIN-93G-MX 39 39 AIN-93-VX ll 11 L—Cystine 3 3 Choline Bitartrate 3 3 Sample Collection: Mouse body weights were measured weekly. After ten weeks of dietary treatments, mice were euthanized. Blood samples from each mouse (approximately 300 ILL) were collected immediately from the heart using a needle and syringe and transferred to microcentrifuge tubes and held on ice. Once the clot formed, the samples were centrifuged and the serum was carefully aspirated into a fresh tube and stored at —80 °C. A small piece of liver was sampled (usually at the lower edge of the left lobe) and stored at —80 °C. The entire intestine was removed, opened longitudinally, rinsed with water, pinned on cardboard, and fixed with 10% neutral buffered formalin overnight. A one-centimeter section was cut from the middle of the colon and processed and paraffin-embedded for firrther analyses of immunohistochemistry. The fixed intestinal tissues were stained with 0.3% methylene blue for three minutes to facilitate quantification of adenoma numbers and sizes. 37 Quantification of Intestinal Tumors: Numbers of adenomas in each intestinal section (proximal, middle, and distal small intestine, cecum, and colon) were determined by using a stereo microscope. The dimensions of each adenomas was measured using a transparent grid placed under the specimen. The diameter of each adenoma was estimated to the nearest 0.5 mm. For flat tumors in the small intestine, the diameters were measured in two dimensions (d1, d2), and the Sizes of the adenomas were calculated by using the formula: a = (II * d1 * d2) / 4. For solid tumors in the large intestine, the diameters were measured in three dimensions (d1, d2 and d3), and the spherical volumes were calculated by using the formula: spherical volume = (1r "' d1 * d2 * d3) / 6. Immunohistochemistry: Paraffin-embbed tissue blocks were cut into tissue sections (4 pm thick) and oriented on Poly-L-lysine slides. Tissue sections were adhered to slides by drying at 58°C in a vacuum oven for two hours, and then they were deparaffinized with xylene (2 x 5 min), rehydrated through 100% and 95% ethanol (2 x 2 min each), respectively, then rinsed in nanopure water 3 times. Endogenous peroxidase activity was blocked by immersion in 0.3% hydrogen peroxide for 10 min, followed by washing with buffered solution (TBS or PBS, pH 7.4) 3 times. Antigen retrieval was achieved by using citrate buffer (10 mM sodium citrate) at 95°C for 20 minutes followed by cooling of 20 minutes. After thoroughly washing with TBS, the slides were covered with capillary-action slide clips and mounted on Shandon IHC staining racks (Shandon Immunohistochemical System, Thermo Electron Corporation). Specific immunohistochemistry staining procedures were conducted for PCNA and Ki67 antigens. Reagents were obtained from DakoCytomation (Carpinteria, CA) unless identified otherwise. 38 A. Measurement of PCNA: Tissue sections were incubated with an anti-PCNA antibody (PC10, Novcastra, UK; 1:100 dilution with 2% BSA in TBS) overnight at 4°C. Slides were rinsed twice with TBS and then incubated with Link Antibody (goat anti- mouse IgG) (4 drops) for 30 minutes. Slides were rinsed twice with TBS and then incubated with strepavidin-peroxidase conjugate (4 drops) for another 30 minutes. Slides were then washed with TBS twice and incubated in 0.05% DAB (3,3'—Diaminobenzidine; Sigma) for 7-10 minutes. Slides were then rinsed and counterstained with Mayer's hematoxylin (Sigma) for 15 seconds, rinsed and blued in 0.3% ammonia for 5 seconds. Finally, slides were coverslipped using Faramount aqueous mounting media. Ten full-length crypts of each colon were examined to determine labeling indices (all the crypts appeared to have normal cryptal structure, and crypts adjacent to adenomas were excluded). The number and the position of PCNA-stained nuclei in each crypt column were recorded. PCNA labeling index (LI) was determined as the percentage of cells positively stained in each colon hemi-crypt. The colon hemi-crypt was also divided into three sections with equal numbers of cells: the bottom third, the middle third and the top third. The labeling index also was calculated for each section as the percentage of cells positively stained in each section. The proliferative zone was calculated using the formula: proliferative zone = position of highest labeled cell in crypt / crypt height. B. Measurement of Ki67: The tissue sections were incubated with an anti-Ki67 antibody (NCL-Ki67, Novcastra, UK; 1:100 dilution with 2% BSA in TBS) overnight at 4°C. All the subsequent steps were the same as that used for PCNA measurement. 39 C. Measurement of Apoptosis: The apoptosis index in colonic epithelial cells was measured by the terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick end-labeling (TUNEL) assay using the in situ cell death detection kit from Roche Molecular Biochemicals (Nonnenwald, Germany), which labels DNA strand breaks produced during apoptosis. Briefly, tissue sections (4 pm) were de-paraffinized with xylene, rehydrated as described before and washed with phosphate-buffered saline (PBS). Antigen retrieval was achieved by using citrate buffer (10 mM sodium citrate) at 95°C for 20 minutes followed by cooling of 20 minutes. Slides were washed twice in PBS and treated with proteinase K (2 jig/ml in 10 mM Tris—HCI) for 15 min at room temperature. Slides were then rinsed twice with PBS, incubated with 3% H202 for 10 min at room temperature, and again rinsed twice with PBS. Slides were then covered with a slide cover slip and mounted on Shandon IHC staining racks (Thenno Electron Corporation). Slides were exposed to 2% bovine serum albumin for 20 min to mask non-specific antigens. TUNEL reaction mixture (50 pl) consisting of terminal deoxynucleotidyl transferase and the nucleotide were added (the original Enzyme Solution in vial l was diluted with 2 times volume of TUNEL Dilution Buffer, then 5 ul of the diluted enzyme were mixed with 45 ul of the Label Solution to obtain the 50 ul working TUNEL reaction mixture). The slides were incubated for 60 min at room temperature in dark. Slides were then rinsed three times with PBS, incubated for 30 min at 37°C with converter-POD peroxidase-conjugated antibody and rinsed three times with PBS. Slides were then incubated for 10 min at room temperature with 100 pl of ABC chromogen substrate solution, counterstained with hematoxylin for 15 seconds, rinsed in distilled water, blued with 0.3% ammonia water, and coverslipped using Faramount aqueous mounting media. 40 Cells having red stained nuclei were identified as TUNEL positive. Ten intact intestinal crypts were scored for each animal, and the apoptotic index was calculated as the number of stained cells in each crypt divided by the total number of epithelial cells in each crypt. Measurement of Glucose Concentrations in Serum: Serum glucose concentrations were measured using the QuantiChrom Glucose Assay Kit (DIGL-200, BioAssay System, Hayward, CA) by following the manufacturer’s instructions. Briefly, 5 pl glucose standards and samples were transferred to appropriately labeled tubes. 500 pl Glucose Assay Reagent was added to each tube. The tubes were closed tightly and thoroughly mixed. Samples were then heated in a boiling water bath for 8 min and cooled in cold water bath for 4 min. Duplicate samples (200 pl) from each sample were transferred into a clear-bottom 96-well plate. Optical density was measured at 570 nm using a microplate reader, and the absorbance data were analyzed using SOFTmax PRO (Molecular Devices Corporation, Sunnyvale CA). Measurement of Insulin Concentrations in Serum: Concentrations of insulin in blood serum were measured using the Mercodia Mouse Insulin ELISA kit (Mercodia AB, Uppsala, Sweden) by following the manufacturer’s instructions. Briefly, 25 pl each of the standards and samples were added to a 96-well plate coated with anti-insulin antibodies. Enzyme conjugate (50pl) was added to each well and plates were incubated on a shaker for 2 hours at room temperature. Plates were then washed 6 times with an automatic plate washer using the wash buffer solution. The chromogenic substrate 3,3',5,5'- tetramethylbenzidine (TMB; 200 pl) was added to each well and plates were incubated for 15 min, at which time 50 pl stop solution was added. Optical density of each well 41 was measured at 450 nm using a microplate reader, and the absorbance data were analyzed using SOFTmax PRO (Molecular Devices Corporation, Sunnyvale CA). Quantitative Real Time PCR Analysis of Hepatic mRNA Expression of IGF- I, IGF-II, IGFBPl and IGFBP3: A. Total RNA Extraction: Total RNA from liver samples was extracted using Trizol reagent according to the manufacture’s instructions. Briefly, liver samples (200- 300 mg) were suspended in 1 ml Trizol, homogenized and incubated 10 minutes at room temperature. chloroform (200 pl) was added to the suspension and phases were separated by centrifugation for 10 minutes (12,000 x g) at 4°C. The aqueous phase was aspirated into a new tube and RNA was precipitated with 0.5 ml isopropyl alcohol. The total RNA pellets were washed with 75% ethanol and resuspended into RNAase free H2O. The RNA concentrations were determined by spectrophotometry, and RNA integrity was monitored by agarose gel electrophoresis. B. Primer Design: Consensus sequences for genes of interest were obtained from the NCBI website. The Primer3 program (http://www-genome.wi.mit.edu/cgi- bin/primer/primer3_www.cgi) was used to design the primers for each gene of interest. General conditions used in the Primer3 program included: primer size 15-27 hp, primer melting temperature 57°-63°C, and product size 150-250 bp. The resulting primers (Table 2) were synthesized in the Genomic Technology Support Facility in the Department of Biochemistry at Michigan State University. 42 Table 2. List of primers used for real time PCR for gene expression in experiment 1. Gene Forward primer (5’-3’) Reverse primer (5’-3’) Amplicon Name length (bp) B-ACTIN GCTACAGC'ITCACCACC TCTCCAGGGAGGAAGAG 123 ACA GAT IGF 1 CTACCAAAATGACCGCA CACGAACTGAAGAGCAT 126 CCT CCA IGF-II CCCTCAGCAAGTGCCTA TTAGGGTGCCTCGAGAT 121 AAG GIT IGFBPI AGCCCAGAGATGACAGA GITGGGCTGCAGCTAAT 199 GGA CTC IGFBP3 TGTI'ITCTGGTCCAGCC CAAGCCACTCCTCTTTC 122 TCT CTG *Abbreviations: insulin-like growth factor I (IGF-I), insulin-like growth factor II (IGF- II), insulin-like growth factor binding protein 1 (IGFBPI), insulin-like grth factor binding protein 3 (IGFBP3). C. Synthesis of cDNA by Reverse Transcription: cDNA was synthesized from total RNA using the Superscript II system (Invitrogen). Each reaction tube contained 1.0 pg total RNA and 1.0 pl of 0.02 mM anchored oligo dTl8VN (primers having 18 thymidine residues with one G, C or A residue as the anchor at the 3' end to ensure binding at the beginning of the mRNA message) and water up to a final volume of 12 pl. The reaction solution was mixed and incubated at 70°C for 10 minutes to denature the templates, then held on ice for 1 minute to anneal primers to templates. Then, the reaction tube was pulse-spinned, and 8 pl of master mix (containing 4 pl of 5x first strand buffer, 2 pl of 0.1 M DTT, 1 pl of 10 mM dNTPs, and 1 pl of Superscript II — Invitrogen) was added to each reaction. The reaction solution was mixed and incubated for 2 hours at 42 °C, then inactivated by heating for 15 minutes at 75 °C. The reverse-transcription products were stored at —20 °C until use. 43 D. Quantitative Real time PCR: Preparation of Standards: Prior to conducting quantitative RT-PCR, 1 pl of cDNA mixture containing cDNAs from several samples in different treatment groups was amplified through real-time PCR using primers of the gene of interest (Table 2). Real time PCR assays were conducted using SYBR Green PCR Core Reagents (Perkin Elmer/ABI), using the MicroAmp Optical 96-well reaction plate. The reaction mixture (25 pl final volume) contained 1 pl cDNA reverse-transcribed from 1 pg total RNA, 0.12 mM forward and reverse primers, 2.5 pl of 10x SYBR Green Reaction Buffer, 3 mM MgCl2, 0.2 mM dNTPs, and 0.026 units of Taq polymerase (Perkin Elmer/ABI). PCR reactions were performed in an A81 7700 system with the following cycling conditions: 10 minutes at 95°C, followed by 40 cycles of 15 seconds at 95°C and 1 minute at 60°C. The PCR products were purified using Qiagen PCR purification kits (Qiagen Inc., Valencia, CA) following the manufacturer’s instructions. DNA was eluted in 30 pl elution buffer in the final step of purification, and concentrations of the final PCR products were measured by spectrophotometery. A sample of PCR products were used to run electrophoresis on a 1% agarose gel containing ethidium bromide (4 pl of 10 mg/ml stock per 100 ml of agarose) to verify that each amplicon was of expected size with no non-specific products. The molecular weight of the PCR product for each gene of interest was calculated using the formula: molecular weight (g/mol) = 650 * length of amplicon. The number of copies/ pl was calculated by using the following formula: copy Number (per pl) = (concentration /molecular weight) * 6.023 * 10”. PCR products were each diluted to a copy number of 10l0 and serial dilutions from lO'Oto 101 were created for each product. 44 Quantitative Real Time PCR of Samples: Quantitative real time PCR was carried out by running PCR with 1 pl of each dilution and lpl cDNA of unknown samples on the same plate at the same time. The PCR mixtures and setup conditions were the same as used previously. The quantification of the starting quantity of a specific cDNA for a specific gene in an unknown sample was performed using a standard curve generated by using known dilutions of the corresponding DNA product. For each dilution, the ABI-Prism 7700 software generated a real-time amplification curve constructed by relating the fluorescence signal intensity to the cycle number. The standard curve was then generated on the basis of the linear relationship existing between the Ct value (cycle threshold; corresponding to the cycle number at which a significant increase in the fluorescence Signal was first detected) and the logarithm of the starting quantity. The levels of mRNA for genes of interest were expressed as a ratio between its own expression and that of an internal control gene (beta-actin expression), which was referred as normalized expression. Statistical Analyses: Data were analyzed using the General Linear Models procedure of SAS (Version 8.2). Least square means procedures were used for all the measurements. When significant dietary effects were detected (F-test Significant at P < 0.05), the Least Significant Difference method was used to compare appropriate means. Differences between means were declared significant at P S 0.05. The colon adenoma incidence of mice consuming diets containing sucrose or cornstarch was analyzed using the Frequency procedure of SAS (Version 8.2). 45 RESULTS Intestinal Tumorigenesis: At the end of experiment, twenty-seven and twenty- one APCMin mice were identified using a PCR-based genotyping method (Jacoby et al., 1996) in the sucrose and cornstarch groups, respectively. Mice consuming the sucrose-based diet gained significantly more weight than mice consuming the cornstarch-based diet during the experiment (Figure 5). Although male mice gained more weight than female mice, there was no significant interaction between gender and diet. Based on visual observations during necropsy, much of the observed difference in body weight for the two dietary treatments likely was due to increased abdominal fat deposition in mice consuming the sucrose diet. Dietary carbohydrate source influenced the numbers and average sizes of small intestinal adenomas. These effects were observed exclusively in the proximal one-third of the small intestine. APCMin mice fed sucrose had significantly greater numbers of adenomas in the proximal third of the small intestine than mice fed cornstarch (Figure 6; 21.9 vs. 13.1, P <0.001). However, the average size of adenomas was larger in comstarch-fed mice (Figure 7; 2.1 vs. 1.1 mmz, P <0.01). Overall, there was no difference in the total adenoma burden between the two groups (Figure 8). The total number of adenomas in the small intestine tended to be greater in sucrose-fed mice (Table 3, 72 vs. 60, P <0.07). The colonic adenoma incidence was 56% in APCMin mice fed the sucrose-based diet compared with 43% in APCMin mice fed the comstarch-based diet. However this difference was not statistically significant. Among the tumor-bearing mice, the numbers and sizes of adenomas in colon were numerically greater in mice fed sucrose compared 46 with those fed cornstarch, however these difference did not reach statistical significance (Table 4). 28- 264 24- 22- 20- 18- 16- 14- 12- + Sucrose + Cornstarch Bodyweight (g) 10 I I I 1 I I t j I I j 012345678910 Treatment weeks Figure 5. Weekly body weights of APCMin mice consuming diets containing sucrose or cornstarch as the primary carbohydrate source. Mice consuming sucrose-based diets gained significantly more body weight compared with mice consuming comstarch-based diets (P <0.05). 47 40- lSucrose Diet 35_ I] Cornstarch Diet is 30- a ._..._. g 25- ” 20- E b o 15' 5 3 10" 5- Proximal Small Medial Small Distal Small Intestine Intestine Intestine Figure 6. Numbers of small intestinal adenomas in different small intestinal regions of APCMm mice consuming diets containing sucrose or cornstarch as the primary carbohydrate source. * All APCMm mice had small intestinal adenomas. 1" Significant diet effect (P < 0.01). 48 2.5~ b ISucrose a“ __ DCornstarch E 2~ E o .5 1.5- (D a a E 1- ....... c a: 'a < 0.5- a: m E .. ., _ g V ..... . . < Proximal Small Medial Small Distal Small Intestine Intestine Intestine Figure 7. Average sizes of small intestinal adenomas (mmz) in different small intestinal regions of APCMm mice consuming diets containing sucrose or cornstarch as the primary carbohydrate source. “‘b Significant diet effect (P < 0.01). 49 El Sucrose D Cornstarchl r E E c on 1: h :I In «I E o C o 'o < Distal Small Intestine Intestine Intestine Figure 8. Total burden of small intestinal adenomas (mmz) in different small intestinal regions of APCMm mice consuming diets containing sucrose or corn starch as the primary carbohydrate source. * No significant diet effects were detected. 50 Table 3. Adenoma incidence, average numbers of adenomas per mouse, total adenomaburden per mouse and average size of adenomas in the small intestine of APCM‘" mice consuming diets based on differing carbohydrate sources in experiment 1 (means i standard errors). Parameter Sucrose (n=27) Cornstarch (n=21) For entire small intestine: Incidence of small intestinal adenomas 100% 100% Adenoma number 72.2 i 2.4a 59.7 :t 5.1b Total adenoma burden (mmz) 77.3 :I: 7.3 69.3 i 8.5 Average size (mmz) 1.02 i 0.06 1.17 i 0.07 a,b Diet effect on small intestinal adenoma number (P = 0.07) Table 4. Adenoma incidence, average numbers of adenomas per mouse, total _ adenoma burden per mouse and average Size of adenomas in the colon of APCMln mice consuming diets based on differing carbohydrate sources in experiment 1 (means a: standard errors). Parameter Sucrose (n=27) Cornstarch (n=21) For all APCMin mice: 15/27 9/21 Incidence of colon adenomas 56% 43% Adenoma number 0.66 i 0.14 0.52 3: 0.16 Total adenoma burden (mm3) 6.12 :h 2.00 3.51:1: 2.00 Average Size (mm3) 4.63 :t 1.17 3.27 d: 1.36 For adenoma-bearing mice only: Adenoma number 1.20 i 0.14 1.25 :1: 0.19 Total adenoma burden (mm3) 11,09 :t 2.72 8.38 i 3.72 Average size (mm3) 8.36 :t 1.62 7.75 :l: 2.22 * No significant dietary treatments were detected. 51 Colon Crypt Cell kinetics: Ki-67 antigen labeling was used as a surrogate marker for cell proliferation in this study. Mice consuming sucrose-based diets had significantly greater total crypt Ki-67 labeling index compared to mice consuming cornstarch (Table 5). Crypt height and Ki67 proliferative zone were not influenced by dietary carbohydrate source. Total crypt PCNA labeling index and proliferative zone (Table 6) were not influenced by diet in this experiment. Dietary carbohydrate sources did not influence programmed cell death in colon after lO-weeks of dietary treatment (Table 7). Essentially all programmed cell death occurred in the apical one-third of epithelial cells in colonic crypts. Table 5. Ki67 antigen expression in colon of APCMin mice consuming diets containing different carbohydrate sources (means i standard errors). Parameter Sucrose (n=18) Cornstarch (n=20) Crypt Height (cells) 20.0 d: 0.30 19.9 d: 0.28 Total LI 0.37 d: 0.018a 0.32 a 0.016b Proliferative Zone 0.48 :I: 0.019 0.45 :t 0.018 Bottom 1/3 L1 0.75 i 0.021“‘ 0.66 :1: 0.020b Middle 1/3 Ll 0.28 i 0.037 0.23 :t 0.034 Top 1/3 LI 0.01 i 0.006 0.01 d: 0.006 a,b . Diet effect (P <0.05). 52 Table 6. PCNA antigen expression in colon of APCMin mice consuming diets containing different carbohydrate sources (means :I: standard errors). Parameter Sucrose (n=25) Cornstarch (n=24) Crypt Height (cells) 20.20 i 0.20 20.02 :I: 0.2 Total LI 0.358 t 0.014 0.357 :t 0.014 Proliferative Zone 0.43 i 0.019 0.45 d: 0.019 Bottom 1/3 LI 0.79 i 0.017 0.76 i 0.018 Middle 1/3 LI 0.22 :t 0.026 0.19 :L- 0.026 Top 1/3 LI 0.002 i 0.002 0.003 d: 0.002 ‘ No significant diet effect. Table 7. TUNEL assay results in colon ofAPCMin mice consuming diets containing different carbohydrate sources (means 5: standard errors). Parameter Sucrose (n=18) Cornstarch (n=20) Crypt Height (cells) 19.8 i: 0.14 19.6 i 0.12 Total LI 0.008 d: 0.001 0.008 :t 0.001 Bottom 1/3 LI 0.000i 0.000 0.000 :t 0.000 Middle 1/3 L1 0.000 :t 0.000 0.000 i 0.000 Top 1/3 LI 0.023 :t 0.005 0.026 d: 0.004 ' No significant diet effect. 53 Concentrations of Glucose and Insulin in Serum: Mice consuming high- sucrose diets had significantly greater serum glucose concentrations compared to mice consuming cornstarch (10.09 :I: 0.57 vs. 8.08 at 0.57 mM, P < 0.05; Figure 9). Mice consuming sucrose also had higher plasma insulin levels compared to mice consuming cornstarch (262.6 :I: 34.7 vs. 174.4 1 34.3 pmol/L, P = 0.07; Figure 10). These results suggest that mice fed high-sucrose diets developed insulin resistance and hyperinsulinemia. IGF-I, IGF-H, IGFBP] and IGFBP3 mRNA Expression in liver: As shown in Figure 11, IGF-I relative expression in liver of mice consuming sucrose was greater than that observed in mice fed cornstarch (0.166 vs. 0.092, p = 0.05). IGF-II, IGFBPI and IGFBP3 mRNA expressions in liver were not influenced by diet (Table 8). 54 15_ CI Sucrose CI Cornstarch E. a 9 g 101 b O E. M E a 5‘ '5 O .2 a: Figure 9. Glucose concentrations in blood serum of mice consuming diets containing different dietary carbohydrate sources. ‘1’" Significant diet effect (P < 0.05). 55 350- El Sucrose E El Cornstarch '5 300- E a 5 250- .E . 3 200- b .E S 150- d) w 100« 'U 8 E 50 0 Figure 10. Insulin concentrations in blood serum of mice consuming diets containing different dietary carbohydrate sources. a’b Diet effect (P = 0.07). 56 02- El Sucrose g a D Cornstarch .5 1 J 2 0. 5 S I” b .921 0.1- a 2 E’ —. 0.05“ It. 9 Figure 11. Relative expression of IGF-I mRNA in liver of mice consuming diets containing different dietary carbohydrate sources. a,b Significant diet effect (P < 0.05). 57 Table 8. Relative expression of IGF-II, IGFBPl and IGFBP3 mRNA in liver of mice consuming diets containing different carbohydrate sources (means :I: standard errors). Parameter Sucrose (n=18) Cornstarch (n=20) IGF-II 0.016 :t 0.004 0.011 :t 0.004 IGFBP] 0.0014 :1: 0.0003 0.0011 i 0.0003 IGFBP3 0.012 :t 0.007 0.010 t 0.007 * No significant diet effets. 58 DISCUSSION Epidemiological research and results from experiments using carcinogen-induced colon cancer models suggest that high consumption of refined carbohydrates such as sucrose is associated with increased risk of colon cancer. The influence of high-sucrose diets on intestinal adenoma development in APCMin mice has not been reported previously. In the current study, APCMin mice consuming high-sucrose diets had significantly greater numbers of adenomas in the proximal small intestine, and tended to have more adenomas in the entire small intestine, when compared to mice consuming high- comstarch diets. Although all cells in the intestine of APCMin mice carry a mutant copy of APC, only a relatively small number of tumors form in the intestine. This indicates that further somatic events are required before tumors can develop. It has been suggested that the somatic mutation of the wild-type APC allele inherited fiom the unaffected parent is the rate-limiting step in tumor initiation (Kinzler 1996). It is possible that high- sucrose diets increased mutation rates in the small intestine, which resulted in increased rate of somatic mutation of the second allele of APC. Dragsted et al. (2002) reported that increasing sucrose in diets administered to Big Blue rats for 3 weeks [diet sucrose concentrations = 3.4% (control), 6.9%, 13.8%, or 34.5%] resulted in a dose-dependent increase in the mutation frequency at the CH site in the colonic mucosa, which indicates a direct or indirect genotoxic effect of a sucrose-rich diet. In the current study, the cell proliferation rate in the small intestinal epithelium of APCMin mice was not determined due to difficulties in obtaining intact full-length small intestinal crypts for assessment of cell proliferation. However, cell proliferation rate 59 measured by Ki67 labeling in the colon of APCMin mice consuming high-sucrose diets was significantly greater compared to that measured in the colon of mice fed the high- comstarch diet. It is possible that sucrose-rich diets also increased cell proliferation rate in the small intestinal epithlium in this experiment. It is possible that increased cell proliferation in the small intestine of APCMin mice may result in increased mutation rates, and thereby increase adenoma promotion in the small intestine. APCMin mice fed the high-cornstarch diet had a greater average size of proximal small intestinal adenomas when compared with mice fed the high-sucrose diet. It is possible that small intestinal adenomas in APCMin mice compete for blood and nutrient supply, which ultimately might lead to an upper limit on adenoma burden in the small intestine of these mice. Overall, the adenoma burden in the proximal small intestine did not differ significantly for mice consuming sucrose- or comstarch-based diets. In the current study, dietary carbohydrate source only influenced small intestinal adenoma development in the proximal small intestine. The reason for this is not clear. Dietary sucrose and cornstarch are primarily digested and absorbed in the proximal small intestine in APCMin mice. It has been suggested that Short-term consumption of high- sucrose diets may increase oxidative stress in rats (Busserolles et al. 2002), so it may be possible that chronic exposure to high-sucrose diets may contribute to the observed effects on tumorigenesis in proximal small intestine. We are particularly interested in tumorigenesis in the colon of APCMin mice. Although colon tumor incidence was numerically greater in APCMin mice consuming sucrose versus those consuming cornstarch (56% vs. 43%), this difference was not statistically significant. If high dietary sucrose does increase the risk for colon 60 tumorigenesis, there may be two reasons why no significant differences in colon adenomas were observed in this experiment. Firstly, we may have insufficient statistical power to detect small difference in colonic adenoma incidence. Secondly, small intestinal adenomas initiate and progress very rapidly in APCMin mice, which usually cause severe bleeding, anemia and weight loss. APCMin mice often display morbidity around 15 weeks of age. Because the adenomas in the colon of APCMin mice grow at a slower pace, there may not be sufficient time for diets to exert their full impact on colonic tumor development. Dietary effects on early biomarkers associated with increased risk for colon cancer also were determined using immunohistochemistry analyses. Ki67 labeling is considered a valid biomarker of cell proliferation (Gerdes et al. 1990, 1991; Hall et al. 1990; Holt et al. 1997). In the present study, overall Ki67 labeling index of colonic epithelial cells was significantly greater in APCMin mice consuming sucrose,which indicatsd that consumption of high-sucrose diets increased epithelial cell proliferation in colon crypts. This result is consistent with many other studies in which high-sucrose diets significantly increased epithelial cell proliferation in colon crypts when compared to diets based on cornstarch (Cademi et al. 1991, 1993, 1997). Increased cell proliferation in colon crypts is associated with increased risk for colon cancer (Lipkin et al. 1974. 1984; Bleiberg et al. 1985; Terpstra et al. 1987). Overall, these results indicate that diets containing high sucrose concentrations (versus cornstarch) increased adenoma numbers in proximal small intestine, and also may increase risk for colon tumor development by elevating cell proliferation in colon crypts. Several studies have been conducted to explore possible mechanisms whereby high- 61 sucrose diets increased risk for colon cancer. Cademi et a1. (1996) studied the influence of different dietary carbohydrate sources on colon lumenal environment. Female rats were fed for one month with diets containing different carbohydrate sources. They found that colonic epithelial cell proliferation was significantly increased in rats fed sucrose- based diets when compared to rats fed diets based on glucose, fructose, or cornstarch. However, the effects of carbohydrate source on colonic epithelial cell proliferation were not associated with short-chain fatty acid production or cecal pH (Cademi et al. 1996). Corpet et al. (1998) studied the influence of diets containing different carbohydrate sources on development of insulin resistance and ACF in rats. After receiving an AOM injection, rats were randomly assigned to AIN-76-based diets containing different carbohydrate sources (65% starch, glucose or fi'uctose), and ACF development was measured. The average numbers of ACF per rat were not different for rats fed glucose- or fructose-based diets. Insulin resistance was estimated using indirect insulin resistance markers [FIRI index (blood glucose by insulin), blood triglycerides, and visceral fat]. Indirect insulin resistance markers did not correlate with ACF multiplicity (Corpet et al. 1998). As sucrose and cornstarch both are well digested and the resulting monosaccharides are well absorbed in the small intestine, it is possible that their influence on colon cancer risk may be mediated by factors in the circulation. Animal studies suggest that high intakes of sucrose induce compensatory hyperinsulinemia by causing a decline in insulin sensitivity in the liver and peripheral tissues (Pagliassotti et al., 1995; Storlien et al., 1988; Thorbum et al., 1989; Pagliassotti et al., 1994; Martinez et al., 1994; Pamies-Andreu et al., 1995). Hyperinsulinemia can cause changes in the IGF 62 signaling system, such as increased IGF-I expression in liver, which leads to increased availability of free IGF-I in circulation. Increased exposure of IGF-I receptors to IGF-I in colon epithelium may caused elevated IGF signaling in colonic epithelial cells. Increased IGF Signaling in colonic epithelium is associated with increased colon cancer risk (Giovannucci 2001). In the current study, influence of high-sucrose diets (vs. cornstarch) on weight gain, glucose metabolism, levels of insulin in the serum, and IGF and IGFBP mRNA expressions in liver were further examined. Mice in the sucrose group gained significantly more weight compared with mice consuming cornstarch after lO-weeks of treatment. Sucrose is comprised of two monosaccharides - a glucose and a fructose. Unlike glucose, fructose does not stimulate insulin secretion nor enhance leptin production. Insulin and leptin act as key afferent Signals regulating food intake and body weight. Therefore, dietary fructose may contribute to increased energy intake and weight gain (Elliott et al., 2002). Serum concentrations of glucose and insulin were both greater in mice consuming diets containing sucrose compared to that observed in mice consuming diets containing cornstarch. Animal studies have found that high intakes of sucrose versus cornstarch cause a decline in insulin sensitivity in the liver and later in peripheral tissues as assessed by euglycemic hyperinsulinemic clamps (Pagliassotti et al., 1995; Storlien et al., 1988; Martinez et al., 1994; Pamies-Andreu et al., 1995). Long-term feeding of sucrose-rich diets will cause compensatory hyperinsulinemia. The decreased insulin sensitivity caused by high-sucrose diets is likely related to the fructose component of sucrose (Thorbum et al., 1989; Thresher et al. 2000). 63 It has been proposed that hyperinsulinemia promotes colorectal carcinogenesis and the role of insulin in colorectal cancer is mediated through IGF-I (McKeown-Eyssen 1994; Giovannucci 1995, 2001). To further explore the underlying mechanisms whereby dietary sucrose may modulate colon cancer risk, the influence of high-sucrose diets on IGFS and IGF BPS also were determined. IGF-I and IGF-II in circulation are primarily produced in liver. The relative expression of IGF-I mRNA as measured by quantitative RT-PCR in the liver of mice fed sucrose was significantly greater compared with that in mice fed cornstarch. High insulin concentrations increase the number of growth hormone receptors on hepatic cells, and increased binding of growth hormone to its receptors stimulates the production of IGF-I in liver (Jones et al. 1995; Underwood et al. 1994). The IGF system includes two insulin-like grth factors (IGF-I and IGF-II) which exert their actions on cell growth, differentiation and apoptosis by interacting with IGF-I receptors on the cell membrane. IGF-I and IGF-II are further regulated by a group of specific binding proteins (i.e., IGFBPl through IGFBP6; Stewart et al. 1996; Rajaram et al. 1997). In rodents, IGF-II concentrations are typically low compared with that of IGF-I. In this experiment, the relative expression of IGF-II mRNA in liver was lower compared with that of IGF-I. However, we observed no difference in IGF-II mRNA expression in liver of mice fed sucrose- or comstarch-based diets. IGFBP3 is the major IGF binding protein in the circulation and limits access of both IGF-I and IGF-II to the peripheral tissues. Expression of IGFBPI has been reported to be down-regulated by insulin (Ooi et al., 1992). In the current study, neither IGFBP3 nor IGFBP] mRNA concentrations in liver were influenced by diet. In this experiment, we were not able to measure the concentrations of free IGF-I in the circulation. However, we consider it 64 quite probably that high-sucrose diets would increase circulating IGF-I concentrations due to increased liver IGF-I production. High levels of circulating IGF-I are associated with increased colon cancer risk in humans (Ma et al., 1999; Giovannucci et al., 2000). Wu et al. (2002) demonstrated that the growth of colon adenocarcinomas which were transplanted to the surface of the cecum in control (normal) mice was Significantly greater than that observed in liver- specific IGF-I-deficient (LID) mice, in which serum IGF-I levels are only 25% of that present in normal mice. Intra-peritoneal injections of IGF-I (2 mg/kg body weight) twice daily for 6 weeks increased the serum levels of IGF-I and IGFBP3 in both control and LID mice. Both control and LID mice treated with IGF-I in this manner displayed significantly increased rates of tumor development on the cecum and metastasis to the liver, as compared with saline-injected mice. The number of metastatic nodules in the liver was Significantly greater in control mice when compared with LID mice. These observations indicate that circulating IGF-I levels regulate colon cancer growth and metastasis (Wu et al. 2002). In summary, results of the this study indicate that high-sucrose diets increased small intestinal tumorigenesis in APCMin mice, and this action was associated with increased levels of insulin and IGF in the circulation. The results further indicate that high-sucrose diets increase serum insulin levels and also may increase the levels of free IGF-I in the circulation due to increased production of hepatic IGF-I. Ultimately, this may increase IGF signaling in the peripheral tissues such as intestinal epithelium by increasing the availability of IGF-I to interact with IGF-I receptors present on the intestinal epithelial cells. 65 CHAPTER III. GENE EXPRESSION PROFILING OF INTESTINAL EPITHELIUM IN APCMIN MICE CON SUMIN G DIETS CONTAINING DIFFERENT CARBOHYDRATE SOURCES 66 ABSTRACT Dietary carbohydrate source influences intestinal adenoma development in APCMin mice.. The objective of this study was to further investigate global gene expression alterations in the intestine of APCMin mice caused by APC gene mutations and by dietary carbohydrate source using cDNA microarray analysis. Mice were randomly assigned at 4 weeks of age to one of two modified AIN-93G diets containing either 52.3% sucrose or cornstarch as the sole carbohydrate source. Mice were fed these diets for 10 weeks, at which point they were sacrificed. Total RNA was extracted fi'om epithelial cells scraped from the proximal third of the small intestine. cDNA synthesized using total RNA from small intestine was coupled with fluorescent dyes (Cy3 or Cy5) and hybridized on microarray slides printed with the NIA 15K mouse gene set using a loop experimental design. Expression of 379 genes was significantly different between APCMin mice and wild-type mice. Among these differentially-expressed genes, 109 were annotated and had expression altered by genotype by more than 50%. Many of the genes influenced by APC status were associated with cell growth control and tumorigenesis (APC mutation increased expressions of Clu, Ccnd2, Ccnbl, Btg4, Anxal, Gsptl and decreased expressions of Camkld, septin 2, LatsZ, Dab2, Morf4ll). These observations indicate that APC gene mutations have a broad influence on expression of many genes associated with intestinal tumorigenesis. Expression of 306 genes was significantly influenced by dietary carbohydrate source. Among these genes, 87 were annotated and had expression altered by carbohydrate source by more than 5 0%. The expression patterns of genes altered by dietary carbohydrate source suggest that feeding the high- sucrose diet altered the intestinal expression of genes in the insulin/IGF Signaling 67 pathway (sucrose feeding increased expression of IGF-II and decreased expression of IGFBP3), which may result in elevated IGF signaling in the intestinal epithelium. High- sucrose diets also influenced expressions of genes associated with cell growth control and tumorigenesis (increased expressions of Pena, Csel l, Idb2, Camk2g; decreased expressions of Tial, F ancg, Bmprla, Cul4B). These results yield insights on candidate gene targets for future mechanistic studies to assess the impact of dietary carbohydrates on colon carcinogenesis. 68 INTRODUCTION Modern cDNA microarray technology enables the simultaneous monitoring of mRNA expression of thousands of genes. The application of this technology to conduct gene expression profiling provides a powerful tool to enable researchers to identify candidate genes and pathways that may be involved in regulation of processes such as carcinogenesis (Liefers et al. 2002). APCMin mice have been extensively studied as a model for human colon cancer. APC mutation causes accumulation of B-catenin in the cytosol. These B-catenin proteins can translocate into the nucleus and bind to transcription factors such as T-cell factor (T00 and lymphoid enhancer factor (Let), and increase transcription of a variety of genes having ch/Lef binding sites (Behrens et al., 1996). APC gene mutation is an early event in intestinal tumorigenesis, and APC gene mutations have broad influences on expression of many genes involved in cellular pathways involved in promotion and progression of colon carcinogenesis Dietary carbohydrate concentrations and sources have been demonstrated to impact the risk of colon cancer development. Our previous experiments indicate that dietary carbohydrate source (sucrose versus cornstarch) influences small intestinal adenoma development in APCMin mice. High-sucrose diets also induced hyperinsulinemia and increased hepatic IGF-I mRNA expression in APCMin mice, which suggests an increased availability of insulin and IGF-1 to interact with intestinal IGF-I receptors. However, the potential influence of high-sucrose diets on local gene expression in the intestinal epithelium is not known. 69 The first objective of the current study was to explore broad gene expression changes caused by APC mutations in the small intestinal epithelium of mice using cDNA microarray analysis, and aim to identify gene targets or signaling pathways that are important in colon cancer promotion and progression. The second objective was to investigate early alterations of gene expression patterns (10 weeks of dietary treatment) induced by diets containing different carbohydrate sources (sucrose versus cornstarch). Such pattern alterations may help us to understand the underlying mechanisms whereby dietary sucrose increases intestinal tumorigenesis. . 70 MATERIALS AND METHODS Animals and Diets: APCMin mice used for the study were generated by mating C57BL/6J male APCMin mice with normal C57BL/6J female mice in our breeding colony at Michigan State University. The resulting offspring (about twenty mice with roughly equal numbers of males and females) were assigned to one of two diets containing either sucrose or cornstarch as the sole carbohydrate resource at 4 weeks of age. Among those mice, we expected 50% to be APCMin mice and 50% to have normal APC gene status based on simple Mendelian inheritance. Mice used in this study were genotyped by an allele-specific PCR procedure to identify APCMin progeny (Jacoby et al. 1996). Mice were housed in an animal facility with controlled temperature (about 21- 24°C), humidity (40-70%), and light/dark cycle (12 hours light/ 12 hours dark). Animal care and feeding were conducted with approval of the Michigan State University All- University Committee on Animal Use and Care. Diets used in the experiment were the same as those used in the first experiment (Table 1). Mice were fed these diets ad libitum for ten weeks. Total RNA Extraction: At the end of experiment, mice were euthanized by C02 asphyxia. A sample of liver tissue was collected from each mouse and frozen for confirmatory APCMin genotyping. The entire intestine from each mouse was removed and cut open longitudinally, rinsed clean with RNAase free H20 and sections for RNA isolation were immersed in RNAlater TM (Ambion, Austin, TX) reagent immediately to inactivate RNAase. Epithelial cells from the proximal small intestine and the entire colon from each mouse were scraped separately with a glass slide into tubes containing Trizol reagent (Invitrogen, Carlsbad, Ca), and frozen at -80°C for future RNA extraction. Total 71 RNA from epithelial cells was extracted using Trizol reagent according to the manufacture’s instructions. The RNA concentrations were determined by spectrophotometry, and RNA integrity was monitored by agarose gel electrophoresis. Preparation of the Microarray Slides: Microarray Slides were printed with genes from the National Institute on Aging’s mouse 15K cDNA clone as described by Tanaka et al. (2000). Printing of the slides was carried out at the Plant Research Laboratory at Michigan State University according to the manufacturer’s protocol (Telechem, Sunnyvale, CA). cDNA Synthesis and Fluorescence Labeling of Microarray Probes: Synthesis of fluorescence-labeled cDNA was conducted based on procedures developed by The Institute of Genomic Research (Hegde et al., 2000). Briefly, each RNA sample (20 pg per reaction) extracted from proximal small intestine was denatured and primed with Oligo dT, then reverse-transcribed into cDNA using Superscript II transcriptase (Invitrogen, Carlsbad, CA) in the presence of aminoallyl modified dUTP (Sigma Chemical Company, St. Louis, Mo) (aa-dUTP: dTTP = 2:1) according to manufacture’s instructions. Dye coupling was conducted by incubating cDNA with the monoreactive dyes cyanin 3 (Cy3) or cyanin 5 (Cy5) (Amersham, Buckinghamshire, UK) for 1 hour at room temperature in the dark. Cy3- and Cy5- labeled samples for a given slide were mixed according to the experimental design. Unincorporated dyes were removed using the Qiagen cleanup kit (Quiagen Inc.). Each probe (Cy3 and Cy5 labeled pair) was dried in a speed vacuum and resuspended in pre-hybridization buffer. 72 Microarray Hybridization: before hybridization, microarray slides were pre- treated to remove unbound DNA-molecules and buffer substances. The slides were rinsed in the following steps: five minutes in 0.1% Trition-X 100, two rinses of two minutes each in HCl solution (100 pl of 37% HCl in 1000 ml H20), and ten minutes in 100 mM KCl solution. The microarray slides were then rinsed in boiling H2O for three minutes and washed for one minute in H2O at room temperature. The slides were then treated with 1X QMT Blocking Solution (Quantifoil, Jena, Germany) at 50°C for 15 minutes, and rinsed in H2O for one minute before hybridization. Each probe was hybridized to pre-treated microarray slides according to the experimental design (Figure 12). All of the hybridization steps were conducted in a dark room. Briefly, the dried probes were re-suspended in 50 pl of formamide-based hybridization buffer (28 pl of formamide, 2 pl of 20% SDS, 4 pl of 50X Denhardt’s and 16 pl of 20X SSPE), which was preheated to 42°C. Then, 2 pl of polyA+ (10 pg/pl; Gibco), 2 pl of mouse COT-1 DNA (10 pg/pl; Gibco) and 1 pl of yeast tRNA (10 pg/pl; Gibco) were added. The hybridization solution containing the probe was vortexed lightly and incubated at 42°C for two minutes, and this step was repeated three times. The probe was then denatured at 100°C for three minutes and then held on ice for one minute. The probe was centrifuged at 12000 x g for one minute and was applied to the microarray slide immediately. After adding the probe, each slide was cover-slipped and placed in the hybridization chambers (Coming), in which the slides were incubated at 42°C for 18 to 24 hours. After incubation, the microarray slides were washed clean in dark room. Briefly, the slides were removed from hybridization chambers and placed in slide racks in 73 preheated (42°C) 2X SSC solution containing 0.2% SDS until the cover slips slid off. Slides were then washed in 2X SSC solution containing 0.2% SDS at room temperature for five minutes. Slides were then transferred to a new slide rack and washed in 0.2X SSC solution containing 0.2% SDS for five minutes. They were then transferred into another new slide rack and washed in 0.2X SSC for two minutes, and this step was repeated twice. Finally, the Slides were centrifuged at 1200 rpm to dry and were scanned immediately. Wt Starch 1 _'_—_‘: :7» Wt Sucrose ti M l: :1 l i ii ii Min Starch —fl ——+ Min Sucrose F igure12. Complete loop design for the microarray study. Wt Starch, Wt Sucrose, Min Starch and Min Sucrose represent mice with different APC mutation status and dietary carbohydrate source. Each arrow represents a single microarray slide. In each slide the appropriate samples were labeled with Cy3 (tail of the arrow) and also with Cy5 (head of the arrow). The entire scheme was repeated three times with RNA samples from different mice, which resulted in three independent replicates of the design. 74 Slide Scanning and Data Processing: The microarray slides were scanned using an Affymetrix 425TM scanner (Santa Clara, CA) and the image files were processed using the GenePix Pro 6.0 program (Molecular Devices Corporation, Union City, CA). The resulting GenePix files were imported into the MIDAS (microarray data analysis system) program (www.tigr.org), and the Cy3 and Cy5 signals within each chip were normalized using the Lowess algorithm. The normalized output signals were used for further analyses. Quantitative Real Time PCR (RT-PCR) Analysis: The same RNA samples (n=12) fi'om the proximal small intestine used for cDNA microarrays were also used to confirm gene expression changes with RT-PCR. RT-PCR amplification and data analyses were performed using an ABI Prism 7700 Sequence Detector System (PE Applied Biosystems, Foster City, CA). Consensus sequences for genes of interest were obtained from the NCBI website. The Primer3 program (http://www-genome.wi.mit.edu/cgi-bin/primer/primer3_www.cgi) was used to design the primers for each gene of interest. General conditions used in the Primer3 program included: primer size 15-27 hp, primer melting temperature 57-63°C, and product size 150-250 bp. Primers (Table 9) were synthesized in the Genomic Technology Support Facility in the Department of Biochemistry at Michigan State University. 75 Table 9. List of primers used for confirmatory real time PCR analyses of gene expression. Gene Forward primer (5’-3’) Reverse primer (5’-3’) Amplicon Name length (bp) B-ACTIN GCTACAGCTTCACCACC TCTCCAGGGAGGAAG 123 ACA AGGAT IGFI CTACCAAAATGACCGCA CACGAACTGAAGAGCA 126 CCT TCCA IGF-II CCCTCAGCAAGTGCCTA TTAGGGTGCCTCGAGAT 121 AAG GIT IGFBP3 TGTT'ITCTGGTCCAGCC CAAGCCACTCCTC'ITTC 122 TCT CTG PCNA CCACATTGGAGATGCTG CCGCCTCCTC'ITCTTTA 128 'ITG TCC PIK3C2A CAGTCGAAGCTCTCCTC TTCCAAAATACCAGGA 137 AGC CCTCA CLU CACATGTCTCCAGGCGA AAGGCGGCTT’I'I‘ATTGG 1 33 GTA ATT LTF CAGGGGATCTGGTITCT AGTGCAAGGAGTGCACA 1 3 1 TCA CAG *Abbreviations: insulin-like growth factor binding protein 3 (IGFBP3), insulin-like growth factor II (IGF-II), insulin-like factor I (IGF l), proliferating cell nuclear antigen (PCNA), phosphatidylinositol 3-kinase, C2 domain containing, alpha polypeptide (PIK3C2A), clusterin (Clu), lactotransferrin (LTF). 76 cDNA was synthesized from total RNA using the Superscript II system (Invitrogen). Each reaction tube contained 1.0 pg total RNA and 1.0 pl of 0.02 mM Anchored oligo dT18VN, which were primers having 18 thymidine residues with one G, C or A residue as the anchor at the 3' end to ensure binding at the beginning of the mRNA message, and water up to a final volume of 12 pl. The reaction solution was mixed and incubated at 70°C for 10 minutes to denature the templates, then held on ice for 1 minute to anneal primers to templates. Then, the reaction tube was pulse-spinned, and 8 pl of the master mix was added to each reaction, which included 4 pl of 5X First strand buffer, 2 pl of 0.1 M DTT, 1 pl of 10 mM dNTPs, and 1 pl of Superscript II (Invitrogen). The reaction solution was mixed and incubated for 2 hours at 42 °C, then inactivated for 15 minutes at 75 °C. Finally, the reverse-transcription product was stored at —20 °C until use. Quntitaive real time PCR assays were conducted using SYBR Green PCR Core Reagents (Perkin Elmer/ABI), using the MicroAmp Optical 96-well reaction plate. The reaction mixture (25 pl final volume) contained 1 pl cDNA reverse-transcribed from 1 pg total RNA, 0.12mM forward and reverse primers, 2.5 pl of 10x SYBR Green Reaction Buffer, 3mM MgCl2, 0.2 mM dNTPs, and 0.026 units of Taq polymerase (Perkin Elmer/ABI). PCR reactions were performed in an ABI 7700 system with the following cycling conditions (10 minutes at 95°C, followed by 40 cycles of 15 seconds at 95°C and 1 minute at 60°C). 77 Statistical Analyses: A. cDNA Microarray Analysis: As shown in the microarray hybridization design (Figure 12), each loop was comprised by four mice having different dietary carbohydrate treatment or APC gene status. Four intensities (two Cy3 and two Cy5 intensities) were obtained for individual gene on the microarray chip for each mouse in the loop. For each gene in each animal, the mean and standard deviation were calculated for the four intensity values. Values that were greater than one standard deviation from the mean were eliminated, and the averages of the remaining values were used to represent the gene expression intensities for each gene for that mouse. Three loops of microarray hybridization were conducted separately, which provided three complete biological replicates to allow fiirther statistical analyses. To account for variations between the three loops due to differences in RNA quantity, hybridization and seaming, the individual gene expression intensities for each mouse were further normalized against the median intensity of all gene expression intensities for that mouse using the GeneSpring 6.0 program (Silicon Genetics, Redwood City, CA). Two-way ANOVA analyses were performed using GeneSpring for main effects of genotype (wild-type versus APCM'") and diet (sucrose versus cornstarch) on expression of each gene. Differentially-expressed gene transcripts (P < 0.05) were classified into functional groups based on known gene ontologies and putative fiInctions reported in the literature. Gene ontologies were retrieved from the Rat Genome Database (RGD) maintained at the Medical College of Wisconsin (Milwaukee, Wisconsin; URL: http://rgd.mcw.edu/). 78 To compare APC status effects (APCMin vs. wild-type) on relative gene expression, a ratio for each gene was calculated by dividing the expression in mice carrying APC gene mutations by that observed in wild-type mice. To compare the effect of diets containing different carbohydrate sources (sucrose vs. cornstarch) on relative gene expression, a ratio for each gene was calculated by dividing the gene expression observed in mice consuming sucrose by that observed in mice consuming cornstarch. Because relative expressions of a large number of genes were significantly (P < 0.05) influenced by diet or APC mutation, a cutoff (if up-regulated, ratio 2 1.5; if down-regulated, ratio 5 0.6) was used to identify genes whose expression changes are biologically meaningfiIl and also to reduce the numbers of genes in the list presented in the results. Genes listed in the results include those that were significantly influenced by genotype or diet (P <0.05) and also meet these cutoff conditions. Many of the gene elements corresponding to unknown genes based on the current gene ontology were not reported here even though their expression levels were significantly altered by either APC gene mutation or diet (complete lists of genes whose expressions were Significantly altered by diets or APC gene mutation are attached as Table A1 — Table A5 in the appendix). B. Quantitative Real-Time PCR Analysis: The standard curve method was used to quantitate the mRNA levels. Quantification of the starting quantity of a specific cDNA generated for an unknown sample was performed using a standard curve using known dilutions of the corresponding DNA product. For each dilution, the AbI-Prlsm 7700 software generated a real-time amplification curve constructed by relating the 79 fluorescence signal intensity to the cycle number. The standard curve was then generated on the basis of the linear relationship existing between the Ct value (cycle threshold; corresponding to the cycle number at which a significant increase in the fluorescence signal was first detected) and the logarithm of the starting quantity. The normalized expression of a gene of interest was calculated as a ratio of its own mRNA expression against the mRNA expression of the internal control gene (beta-actin). Normalized expression from RT-PCR analyses were analyzed as a 2 x 2 factorial design (diet, genotype) using the General Linear Models procedure of SAS (SAS Institute, Inc.; Cary, NC, Version 8.0). 80 RESULTS A. Effect of APC gene mutation on gene expression: As shown in table 10 and l 1, we observed that there were 55 genes up-regulated (with ratio 2 1.5) and 54 genes down-regulated (with ratio 3 0.6) in mice which were heterozygous for the APC gene mutation when compared with wild-type mice. When classified into functional categories, these genes included those coding for cell cycle and growth control (up 8, down 5), Signal transduction and receptors (up 10, down 9), transcriptional and translation regulation (up 10, down 11), cell adhesion and cytoskeleton (up 4, down 2), cellular component and structure proteins (up 5, down 3), transport and carrier proteins (up 7, down 12), and metabolism and enzymes (up 11, down 12). Several genes related with the Wnt signaling pathway were Significantly influenced by APC gene status (APCMin vs. wild-type). Wnt4 expression was increased about 2.4 fold in APCMin mice compared with normal mice. Expression of Dab2, which is considered as an antagonist of the Wnt signaling pathway (Prunier et al. 2004; Dote et al. 2005), was down-regulated in APCMin mice. Expression of cyclin-D2, but not that of cyclin-D1, was found to be significantly increased in APCMin mice compared with normal mice. A large group of genes involved in cell growth control and tumorigenesis was differently expressed in APCMin mice and normal mice. The genes overexpressed in APCMin mice included clusterin (Clu), cyclin-D2 (Ccnd2), cyclin-Bl (Ccnbl), B-cell translocation gene 4 (Btg4), annexin A1 (Anxal), G1 to S phase transition 1 (Gsptl) and bone morphogenetic protein receptor, type 1A(Bmpr1a). Genes in this category that 81 were down-regulated in APCMin mice included calcium/calmodulin-dependent protein kinase ID (Camkld), septin 2, large tumor suppressor 2 (Lats2), disabled homolog 2 (Dab2), and mortality factor 4 like 1 (Morf4ll). Genes up-regulated in APCMin mice included those involved in various signal transduction pathways. These genes included protein phosphatase 5 (Ppp5c), Wnt4, mitogen-activated protein kinase 6 (Mapk6), protein tyrosine phosphatase 4a3 (Ptp4a3), T-complex expressed gene 1 (Tcel), Rho guanine nucleotide exchange factor 10 (Arhgefl 0), immunoglobulin-like domain containing receptor 1 (lldrl), endoplasmic reticulum (ER) to nucleus signalling 1 (Eml) and ADP-ribosylation factor-like 3 (Arl3). Genes involved in various signal transduction pathways that were down-regulated in APCMin mice included receptor (calcitonin) activity modifying protein 2 (Ramp2), estrogen receptor 1 (alpha) (Esrl), bisphosphate 3'-nucleotidase 1 (Bpntl), HIV-1 Rev binding protein-like (Hrbl), interleukin l7 receptor (Ill7r), PDZ domain containing 2 (szk2), tumor necrosis factor receptor superfamily, member 19 (Tnfrsf19) and homeodomain interacting protein kinase 3 (Hipk3). Another group of genes regulated differentially by APC gene status were those involved in regulation of transcription and translation. Almost equal numbers of genes in this class were up- or down-regulated in APCMin mice. The genes that were up-regulated in APCMin mice included high mobility group box 3 (ngb3), zinc finger protein 592 (pr592), general transcription factor 11 H, polypeptide l (th2h1), MYB binding protein (P160) 1a (Mybbpla), interferon induced transmembrane protein 3 (Ifitm3) and eukaryotic translation initiation factor 2, subunit 2 (Eif252). The down-regulated genes in 82 APCMin mice include ubinuclein l (Ubnl), transcription factor 19 (ch19), transforming growth factor beta 1 induced transcript 4 (Tgfbli4), thyroid hormone receptor interactor 4 (Trip4), nuclear receptor co-repressor 1 (Ncorl), stem-loop binding protein (Slbp) and replication protein A1 (Rpal). Expressions of several transport proteins were increased in APCMin mice. These included synaptotagmin 11 (Sytl 1), ubiquitously expressed transcript (Uxt), lactotransferrin (Ltf), metallothionein 2 (Mt2), abl-interactor 2 (Ab12), solute carrier family 15, member 4 (Slc15a4) and solute carrier family 11, member 2 (Slc11a2). Genes coding for transporter proteins that were significantly down-regulated in APCMin mice included nucleoporin 62 and 37 (Nup62 and Nup37), sideroflexin 1 (Sfxnl), ADP- ribosylation factor-like 6 interacting protein 5 (Arl6ip5), oxysterol binding protein-like 8 (Osbp18) and F K506 binding protein 10 (Fkbp10). 83 Table 10 Genes up-regulated in small intestinal epithelium of APCMin mice versus wild-type mice. GeneSymbol GeneName APCMin/Wt CloneID Ratio Cell cycle and growth control Clu clusterin 5.6 H3108A04 Btg4 B-cell translocation gene 4 14.9 H3051H08 Ccnbl cyclin B1 2.9 H3 105810 Anxal annexin A1 2.0 H3008H01 Ccnd2 cyclin D2 1.8 H3152D01 Gsptl G1 to S phase transition 1 1.6 H3114F01 Psmd8 proteasome (prosome, macropain) 1.5 H3081H11 26S subunit, non-ATPase, 8 Bmprla bone morphogenetic protein 1.6 H3031E08 receptor, type 1A Signal transduction and receptors Ppp5c protein phosphatase 5, catalytic 1.7 H3158H01 subunit Wnt4 Wingless-related MMTV integration 2.4 H3159E07 Site 4 Tcel T-complex expressed gene 1 2.1 H3138A07 Mapk6 mitogen-activated protein kinase 6 2.0 H3064B03 Ptp4a3 protein tyrosine phosphatase 4a3 1.9 H3088F03 ArhgeflO Rho guanine nucleotide exchange 1.8 H3137A10 factor (GEF) 10 Arhgef3 Rho guanine nucleotide exchange 2.9 H3034F 08 factor (GEF) 3 Ildrl immunoglobulin-like domain 2.3 H3102H08 containing receptor 1 Eml endoplasmic reticulum (ER) to 2.5 H3 144812 nucleus signalling 1 Arl3 ADP-ribosylation factor-like 3 2.6 H3092810 Transcription and translation regulation ngb3 high mobility group box 3 1.8 H3030H10 Jaridla jumonji, AT rich interactive domain 3.5 H3154D09 1A (Rbp2 like) Nhlrc2 NHL repeat containing 2 3.5 H3096B06 pr592 zinc finger protein 592 2.9 H3148F06 84 Table 10 (cont’d) th2hl Mybbpl a Ifitm3 Cpsfl Rpsl 1 Eif252 general transcription factor 11 H, 2.7 polypeptide l MYB binding protein (P160) la 2.3 interferon induced transmembrane 2.3 protein 3 cleavage and polyadenylation 2.4 specific factor 1 ribosomal protein 811 1.5 eukaryotic translation initiation 1.5 factor 2, subunit 2 (beta) Extracellular matrix, cell adhesion and cytoskeleton Was Kif14 Btbd9 Gpcl Was Wiskott-Aldrich syndrome homolog 1.9 (human) kinesin family member 14 1.8 BTB (POZ) domain containing 9 2.0 Glypican 1 2.2 Wiskott-Aldrich syndrome homolog 1.9 (human) Cellular component and structure proteins Psmdl 1 Ap251 Psme4 Rps 1 7 Rp524 proteasome (prosome, macropain) 1.6 26S subunit, non-ATPase, 11 adaptor-related protein complex 2, 1.6 sigma 1 subunit proteasome (prosome, macropain) 1.6 activator subunit 4 ribosomal protein 817 1.5 ribosomal protein 824 1.5 Transport and carrier proteins Sytll Slc15a4 Slc11a2 Uxt Abi2 Ltf Mt2 synaptotagmin 11 3.2 solute carrier family 15, member 4 3.1 solute carrier family 11 (proton- 2.2 coupled divalent metal ion transporters), member 2 ubiquitously expressed transcript 2.0 abl-interactor 2 1.5 Lactotransferrin 2.5 metallothionein 2 1.5 85 H3 142H06 H3008A10 H3107D05 H3150D10 H3010A10 H3009C11 H3020F08 H3049F12 H3142F10 H3144G12 H3020F08 H3112D08 H3102F01 H3 120D03 H3118GO4 H3145D11 H3106E08 H3109B08 H3029B01 H3127Bl l H3024D04 H3087A12 H3013D11 Table 10 (cont’d) Metabolism an enzymes Fbxo6b Sepxl Mat2b Pgm211 Expi B3 gntl Ube2r2 Hprtl Uchll Slk Acbd3 F -box only protein 6b 1.6 selenoprotein X 1 3.7 methionine adenosyltransferase II, 2.2 beta phosphoglucomutase 2-like 1 1.9 extracellular proteinase inhibitor 1.8 UDP-GlcNAczbetaGal beta-1,3-N- 1 .8 acetylglucosaminyltransferase 1 ubiquitin-conjugating enzyme E2R 2 1.7 hypoxanthine guanine 1.5 phosphoribosyl transferase l ubiquitin carboxy-tenninal hydrolase 1.5 L1 STE20-like kinase (yeast) 3.0 acyl-Coenzyme A binding domain 1.6 containing 3 86 H3013A04 H3002E01 H3152D10 H3 0993 1 1 H3097H03 H3002H04 H3008B03 H3152G02 H3059F01 H3 040D06 H3 125 E03 Table 1] Genes down-regulated in small intestinal epithelium of APCMin mice versus wild-type mice. GeneSymbol GeneName APCM‘"/ CloneID Wt Ratio Cell cycle and growth control Camkl d septin 2 LatsZ Dab2 Setdbl Morf4l 1 Stkl 1 calcium/calmodulin-dependent protein kinase ID septin 2 large tumor suppressor 2 Disabled homolog 2 (Drosophila) SET domain, bifurcated 1 mortality factor 4 like 1 serine/threonine kinase 11 Signal transduction and receptors Ramp2 Ppp4r1 Esrl Bpntl Hrbl Il 17r szk2 Tnfrsfl 9 Hipk3 receptor (calcitonin) activity modifying protein 2 0.2 H3104G10 0.6 H3152Ell 0.6 H3112G08 0.6 H3112G10 0.7 H3062Cll 0.5 H3048C04 0.7 H3052C12 0.3 H3 109A04 protein phosphatase 4, regulatory subunit 0.6 H3022D11 1 estrogen receptor 1 (alpha) bisphosphate 3'-nucleotidase 1 HIV-1 Rev binding protein-like interleukin 17 receptor PDZ domain containing 2 tumor necrosis factor receptor superfamily, member 19 homeodomain interacting protein kinase 3 Transcription and translation regulation Ubnl chl9 Tgfbl i4 Trip4 Nabl Ncorl Slbp F em 1 a Twist2 ubinuclein 1 transcription factor 19 transforming growth factor beta 1 induced transcript 4 thyroid hormone receptor interactor 4 Ngfi-A binding protein 1 nuclear receptor co-repressor 1 stem-loop binding protein feminization 1 homolog a (C. elegans) twist homolog 2 (Drosophila) 87 0.6 H3095A05 0.5 H3012G10 0.5 H3116F04 0.6 H3008A03 0.5 H3076F01 0.6 H3076A10 0.2 H3155D07 0.2 H3080D07 0.3 H3074GI 1 0.3 H31 12G06 0.5 H3018D01 0.6 H3056A11 0.6 H3070A10 0.4 H3113A01 0.3 H3030A09 0.6 H31 16C08 Table 1 l (cont’d) Ddx3x DEAD/H (Asp-Glu-Ala-Asp/His) box polypeptide 3, X-linked Rpal replication protein A1 Extraceullar matrix, cell adhesion and cytoskeleton Spg20 spastic paraplegia 20, spartin (Troyer syndrome) homolog (human) Wdrl WD repeat domain 1 Cellular component and structure proteins Fin14 fibroblast growth factor inducible 14 Mpvl 7 Mpv17 transgene, kidney disease mutant Anxa4 Annexin A4 Transport and carrier proteins Nup62 nucleoporin 62 Sfxnl sideroflexin 1 Nup37 nucleoporin 37 Grikl glutamate receptor, ionotropic, kainate 1 SlclSa2 solute carrier family 15 (H+/peptide transporter), member 2 Uncl3c unc-13 homolog C (C. elegans) Slc23a2 solute carrier family 23 (nucleobase transporters), member 2 Arl6ip5 ADP-ribosylation factor-like 6 interacting protein 5 Mttp microsomal triglyceride transfer protein Osbpl8 oxysterol binding protein-like 8 Gcipip GCIP-interacting protein p29 F kbp10 FK506 binding protein 10 Metabolism and enzymes Ndufs7 NADH dehydrogenase (ubiquinone) F e- S protein 7 Spint2 serine protease inhibitor, Kunitz type 2 Reel Ras and a-factor-converting enzyme 1 homolog (S. cerevisiae) Acaa2 acetyl—Coenzyme A acyltransferase 2 (mitochondrial 3-oxoacyl-Coenzyme A thiolase) Asah2 N-acylsphingosine amidohydrolase 2 Senp6 SUMO/sentrin specific protease 6 88 0.6 0.5 0.6 0.5 0.6 0.5 0.6 0.3 0.5 0.6 0.2 0.3 0.3 0.5 0.6 0.6 0.6 0.5 0.6 0.6 0.6 0.5 0.6 0.6 H3068C04 H3030A08 H3016G03 H3024D09 H3068C04 H3 140G06 H3087E01 H3105F08 H3026E07 H3067A12 H3147C06 H3073BO3 H3076H09 H3 1 12E08 H3020G12 H3023G06 H3077F04 H3056C1 1 H3 046A09 H3098A08 H3109G10 H3035F09 H31 12307 H3 1 12H01 H3 049A01 Table l l (cont’d) Helicl Ddhdl Ac02 Hibadh Tpil Cblll Helicase, ATP binding 1 DDHD domain containing 1 aconitase 2, mitochondrial 3-hydroxyisobutyrate dehydrogenase triosephosphate isomerase 1 Casitas B-lineage lymphoma-like 1 0.6 0.6 0.6 0.6 0.6 0.2 H3142C01 H3101Cl l H3146GO6 H3069A10 H3149C10 H3068E07 89 B. Effect of dietary carbohydrates on gene expression: As Shown in Tables 12 and 13, there were 54 genes up-regulated (with ratio 2 1.5) and 33 genes down-regulated (with ratio 3 0.6) when mice consumed high-sucrose versus high-comstarch diets. Based on gene ontology annotations, these genes can be classified into several functional categories, including cell cycle and grth control (up 9, down 5), signal transduction and receptors (up 8, down 4), transcription and translation regulation (up 9, down 2), cell adhesion and cytoskeleton (up 2, down 4), cellular component and structure proteins (up 4, down 3), transport and carrier protein (up 5, down 3), host defense and immunity (down 2), and metabolism and enzymes (up 17, down 10) mRNA expression levels of several genes involved in the insulin-like growth factor (IGF) pathway were significantly changed by diets containing different carbohydrate sources (sucrose vs. cornstarch). Gene expression of insulin-like grth factor 2 (IGF-II) in mice fed sucrose was significantly greater than IGF-II expression in mice fed cornstarch, whereas expression of insulin-like growth factor binding protein 3 (IGFBP3) was significantly greater in mice fed cornstarch. Expressions of several genes down-stream in the IGF signaling pathway also were altered. The mRNA levels of Grb7 were induced in mice consuming sucrose, and Grb7 is a homolog of gene Grb2 in the MAPK pathway. The expression of Pik3c2a, the alpha polypeptide of phosphatidylinositol 3-kinase in the PI3K pathway also was elevated by consuming the high-sucrose diet. These gene expression patterns suggest that consuming high sucrose diets may elevate IGF signaling in intestinal epithelial cells. 90 Several genes whose mRNA expression was up-regulated by diets containing sucrose are positive regulators of the cell cycle, and overexpression of these genes are generally associated with increased cell proliferation and growth. These genes included IGF-II, proliferating cell nuclear antigen (PCNA), calcium/calmodulin-dependent protein kinase II gamma (Camk2g), chromosome segregation l-like (Csell), inhibitor of DNA binding 2 (Idb2), multiple endocrine neoplasia 1 (menl), netrin l (Ntnl) and ring finger protein 2 (ng2). Additionally, several genes whose expression were repressed by sucrose containing diets are negative regulators of the cell cycle, and under-expression of these genes are generally associated with increased cell growth. These genes include IGFBP3, cytotoxic granule-associated RNA binding protein 1 (Tial) and bone morphogenetic protein receptor type 1A (Bmprla). Among the genes involved in signal transduction, expression of several genes related to G-protein signaling were induced by high-sucrose diets, including a G protein- coupled receptor (GprcSc), RAS p21 protein activator 3 (Rasa3), Rap guanine nucleotide exchange factor (GEF) 2 (Rapgef2), regulator of G-protein signaling 12 (Rgle) and guanine nucleotide binding protein (Gnaq). However, expression of Rho-guanine nucleotide exchange factor (Rgnef) was down-regulated in mice consuming sucrose. Expressions of the casein kinase 1(Csnkl g2), phosphatidylinositol 3-kinase(Pik3c2a), and phosphodiesterase 7A were elevated, but sphingomyelin phosphodiesterase 3 (Smpd3), ciliary neurotrophic factor receptor (Cntfr) and a signal sequence receptor (Ssr4) were down-regulated by consumption of sucrose-containing diets. Expressions of several genes coding for transcription factors were induced by feeding sucrose. These genes included lymphoid nuclear protein related to AF4 (Laf4), 91 Mad homolog 3 (Smad3) and nuclear factor I/A (Nfia). Conversely, expressions of T- box 20 (Tbx20) and basic leucine zipper and W2 domains 2 (Bzw2) were repressed by dietary sucrose. A large group of genes whose expressions were altered by dietary carbohydrates, code for metabolic enzymes. Some of the genes whose expression was induced by dietary sucrose are involved in carbohydrate and fatty acid metabolism. These included phosphofructokinase (Pfld), aldolase 1 A (Aldoa), stearoyl-Coenzyme A desaturase 2 (Scd2), dihydrolipoamide S-acetyltransferase (Dlat), NADH dehydrogenase (ubiquinone) 1 alpha subcomplex 10 (NdufalO),and aldehyde reductase (aldose reductase)-like 6 (Aldrl6). On the other hand, expression of camitine palmitoyltransferase 1a(Cpt1a), and phospholipid scramblase 3 (Plscr3) were down-regulated. Fibronectin 1 (F ml) and defensin related cryptins (chr), which are involved in host defense and immunity, were found to be down-regulated in mice fed high-sucrose diets. Expression of genes involved in cell adhesion or cytoskeletal structure was also altered by diet. Expression of catenin alpha 1 (Catnal) and thymosin, beta 10 (TmsblO) were induced, whereas, adenylate cyclase-associated protein 1 (Capl), shroom (Shrm), stathmin 1 (Stmnl) and reticulon 4 (Rtn4) were repressed by feeding high-sucrose diet. Other groups of genes regulated by dietary carbohydrates are genes coding for cellular components, and transporter or carrier proteins. C. Interactions of APC gene status and dietary carbohydrate source on eXpression of genes: Table 14 lists genes for which a significant interaction between 92 APC gene status and dietary carbohydrate source was detected. These genes included Rgle (regulator of G protein signaling), Eif356ip (ukaryotic translation initiation factor 3, subunit 6 interacting protein), Cacybp (Calcium binding protein) and several other proteins belonging to cellular component and enzyme categorizes. 93 Table 12. Genes up-regulated in small intestinal epithelial cells of mice consuming diets based on sucrose versus cornstarch. Gene Gene Name Sucrose vs. Clone ID Symbol Cornstarch Cell cycle and growth control Ntnl netrin l 1.5 H3019E06 Csell chromosome segregation l-like 1.7 H3029A05 Igf2 insulin-like growth factor 2 1.5 H3126A04 Menl multiple endocrine neoplasia l 2.2 H3134F11 Pcna proliferating cell nuclear antigen 2.7 H3021F12 Camk2g calcium/calmodulin -dependent 3.6 H3147D07 protein kinase II gamma Rnf2 ring finger protein 2 2.2 H3001812 Idb2 inhibitor of DNA binding 2 3.3 H3118H06 GRB7 Growth factor receptor bound 1.6 H3 006F12 protein 7 Signal transduction and receptors Rasa3 RAS p21 protein activator 3 3.1 H3054E01 Gprc5c G protein-coupled receptor, family 2.9 H3076D03 C, group 5, member C Rapgef2 Rap guanine nucleotide exchange 2.6 H3054804 factor (GEF) 2 Rg312 regulator of G-protein signaling 12 2.2 H3076F 12 Pik3c2a phosphatidylinositol 3-kinase, C2 1.7 H3124D09 domain containing, alpha polypeptide Gnaq guanine nucleotide binding 1.6 H3147806 protein, alpha q polypeptide Csnkl g2 casein kinase 1, gamma 2 2.5 H3119H06 Pde7a phosphodiesterase 7A 1.7 H3085D03 Transcription and translation regulation Laf4 lymphoid nuclear protein related to 1.8 H3093D02 AF4 Smad3 MAD homolog 3 (Drosophila) 3.1 H3149G02 Nfia nuclear factor VA 1.4 H3062812 Nolcl nucleolar and coiled-body 1.9 H3001E10 phosphoprotein l Gcipip GCIP-interacting protein p29 1.6 H3056C11 Pscd3 pleckstrin homology, Sec7 and 3.2 H3095H05 coiled-coil domains 3 94 Table 12 (cont’s) Prdm2 Rbm22 PR domain containing 2, with ZNF 2.5 domain RNA binding motif protein 22 2.2 AI317223 expressed sequence AI317223 1.5 Cell adhesion and cytoskeleton Tmsb l 0 Catnal thymosin, beta 10 1.9 Catenin alpha 1 1.6 Cellular component and structural proteins Srp68 Dibdl an Tbc1d15 signal recognition particle 68 5.3 disrupted in bipolar disorder 1 3.4 homolog (human) tenascin R 2.5 TBCl domain family, member 15 1.6 Transport and carrier proteins Kcnhl Eif4enif1 Timm23 thdl Uqcrb potassium voltage-gated channel, 2.0 subfamily H (eag-related), member 1 eukaryotic translation initiation 2.5 factor 48 nuclear import factor 1 translocase of inner mitochondrial 3.5 membrane 23 homolog (yeast) EF hand domain containing 1 2.1 ubiquinol-cytochrome c reductase 2.1 binding protein Metabolism and enzymes Aldrl6 aldehyde reductase (aldose 4.1 reductase)-like 6 PCYTlA Phosphate cytidylyltransferase 1, 2.1 DLAT Tatdn 1 th40 Ndufal 0 Rps6kal choline, alpha isoform Dihydrolipoamide S- 2.0 acetyltransferase TatD DNase domain containing 1 3.5 DEAH (Asp-Glu-Ala-His) box 2.9 polypeptide 40 NADH dehydrogenase 2.8 (ubiquinone) 1 alpha subcomplex 10 ribosomal protein S6 kinase 1.9 polypeptide 1 95 H3050F05 H3082D03 H3098F12 H3001H10 H3047H12 H3132E08 H3023E04 H3053F08 H3076810 H3033GO9 H3 099A03 H3028F10 H3077C06 H3 107F05 H305 1 E06 H3145805 H3133G11 H3086D07 H3159F10 H3041 A02 H3 002D07 Aldoa aldolase 1, A isoform 1.9 H3031D03 Table 12 (cont’d) Agps alkylglycerone phosphate synthase 1.8 H3031802 Tbkl TANK-binding kinase 1 1.5 H3084809 Pcytla phosphate cytidylyltransferase 1, 2.1 H3145805 choline, alpha isoform Hacel HECT domain and ankyrin repeat 1.7 H3079F02 containing, E3 ubiquitin protein ligase 1 Smpdl3b sphingomyelin phosphodiesterase, 1.7 H3014H09 acid-like 3B Pfkl phosphofi'uctokinase, liver, B-type 1.6 H3025D11 Dlat dihydrolipoamide S- 2.0 H3133G11 acetyltransferase (E2 component of pyruvate dehydrogenase complex) Elovl6 ELOVL family member 6, 2.1 H3077806 elongation of long chain fatty acids (yeast) Scd2 stearoyl-Coenzyme A desaturase 2 3.7 H3025803 96 Table 13 Genes down-regulated in small intestinal epithelial cells of mice consuming diets based on sucrose versus cornstarch. Gene Symbol Gene Name Sucrose vs. Clone ID Cornstarch Cell cycle and growth control Cul4b cullin 48 0.4 H3043809 Tial cytotoxic granule-associated RNA 0.5 H3120F02 binding protein 1 Igfbp3 insulin-like growth factor binding 0.3 H3154G07 protein 3 Fancg Fanconi anemia, complementation 0.4 H3106EO9 group G Bmprla bone morphogenetic protein 0.6 H3031E08 receptor, type 1A Signal transduction and receptors Smpd3 sphingomyelin phosphodiesterase 3, 0.4 H3007A08 neutral ref Rho-guanine nucleotide exchange 0.5 H3139A08 factor Cntfr ciliary neurotrophic factor receptor 0.4 H3075F09 Ssr4 signal sequence receptor, delta 0.5 H3039A08 Transcription and translation regulation Tbx20 T-box 20 0.3 H3040G02 Bzw2 basic leucine zipper and W2 0.3 H3018ElO domains 2 Cell adhesion and cytoskeleton Capl CAP, adenylate cyclase-associated 0.3 H3013E04 protein 1 (yeast) Shrm Shroom 0.5 H3039A02 Stmnl stathmin 1 0.4 H3122C07 Rtn4 reticulon 4 0.6 H3093EOl Cellular component and structural proteins Surf4 surfeit gene 4 0.3 H3014C08 Nduf58 NADH dehydrogenase (ubiquinone) 0.5 H3021805 F e-S protein 8 Rpl6 ribosomal protein L6 0.5 H3144E09 97 Table 13 (cont’d) Transport and carrier proteins Cog8 component of oligomeric golgi 0.4 H3059C05 complex 8 Hba-al hemoglobin alpha, adult chain 1 0.4 H3125H07 Nqo3a2 NAD(P)H:quinone oxidoreductase 0.5 H3043H09 type 3, polypeptide A2 Metabolism and enzymes Cptla camitine palmitoyltransferase 1a, 0.6 H3140A09 liver Plscr3 phospholipid scramblase 3 0.4 H3031809 Aurkc . aurora kinase C 0.1 H3053G07 Pdir protein disulfide isomerase-related 0.3 H3093809 Rcel Ras and a-factor-converting enzyme 0.3 H3035F09 l homolog (S. cerevisiae) Maoa monoamine oxidase A 0.5 H3030809 Xrn2 5'-3' exoribonuclease 2 0.5 H3002C12 Cryzll crystallin, zeta (quinone reductase)- 0.6 H3123G05 like 1 Pgm3 phosphoglucomutase 3 0.3 H3038E09 _Pgm211 phosphoglucomutase 2-like 1 0.4 H3099811 Host defense and immunity Defer defensin related cryptdins 0.3 H3083C07 Fnl fibronectin l 0.3 H3116A10 98 Table 14. Genes whose expressions were altered differently by APC genotype and dietary sucrose Gene Symbol Gene Name M_C w_C M_S w_s Clone ID Signal transduction and receptors Rg512 Regulator of G protein signaling Trancription and translation regulation Eif356ip eukaryotic translation initiation factor 3, subunit 6 interacting protein Transport and carrier proteins Cacybp Calcium binding protein CD28P2 CD2 antigen (cytoplasmic tail) binding protein 2 Abcf ATP binding cassette, sub family Cellular component and structure proteins Psmb7 proteasome (prosome, macropain) subunit, beta type 7 Rga recombination activating gene 1 gene activation Sept2 septin 2 Metabolism and Enzyme Gmppb GDP mannose pyrophosphorylase Clk3 CDC-like kinase 3 Ppp5c Serine/threonine protein phosphatase 5 Ppppl r12a Myosin phosphatase 0.0 0.3 0.3 0.5 1.5 10.0 18.2 18.2 17.9 1.0 0.9 1.1 0.2 0.2 0.2 0.1 0.8 0.6 0.5 0.8 0.7 0.4 2.3 1.5 6.1 2.5 2.0 2.9 0.8 0.9 1.3 1.7 1.1 1.3 0.8 0.8 0.6 5.3 5.9 4.5 0.5 0.6 0.7 11.9 12.1 5.6 H3076F 12 H3025A05 H3147H06 H3106C05 H3031801 H3008G03 H3121C06 H3152E11 H3003E10 H3154809 H3158H01 H3084802 1Data represent normalized mean intensity values of mRNA expression for the corresponding gene in the treatment groups: M_C (APCMm mouse consuming cornstarch), W_C (wild-type mouse consuming cornstarch), M_S (APCMln mouse consuming sucrose), W_S (wild-type mouse consuming sucrose). 99 RT-PCR Confirmatory Analysis of Gene Changes: For APC status effects on gene expression, the mRNA expression of two selected genes (clusterin and lactotransferrin) was further confirmed by RT—PCR using the same RNA samples as in the microarray analysis. Expression of both genes was significantly increased in the proximal small intestinal epithelium of APCMin mice compared with wild-type mice (Table 15). Furthermore, expression of both clusterin and lactotransferrin was also significantly induced in the colon of APCMm mice (Table 16). For effects of dietary carbohydrate source on gene expression, the mRNA expression of five selected genes (IGF-II, IGFBP3, PCNA, PIK3C2A, IGF l) was further confirmed using the same RNA samples from the proximal small intestine by RT-PCR. Although IGF-I expression was not significantly changed by carbohydrate source in the cDNA microarray analysis, we considered it important to further test if IGF-I mRNA expression was altered by using RT-PCR. RT-PCR analysis confirmed that expression of PCNA was significantly (P = 0.05) up-regulated in the small intestinal epithelium of mice fed sucrose compared with those fed cornstarch (Table 17). mRNA expression of IGF 1, IGF-II and PIK3C2A tended to increase, and expression of IGF 8P3 tended to decrease in the proximal small intestine of mice consuming sucrose versus those in mice consuming cornstarch. Although these differences did not reach statistical significance, they were correlated with the results of cDNA microarry analysis. RT-PCR analysis also was performed using total RNA extracted from the colon of the same mice. mRNA expression of IGF-II was significantly up-regulated in the colon epithelium of mice fed dietary sucrose (P < 0.05; Table 18). Expressions of PCNA and 100 PIK3C2A also tended to be greater in mice consuming sucrose diets. However, expression pattern changes of IGFBP3 and IGF-1 caused by dietary carbohydrate source were different in colon compared with that in proximal small intestine. Expression of IGFBP3 in the colon of mice consuming sucrose-based diets tended to be greater than that in mice consuming cornstarch-based diets, whereas, expression of IGF-I in the colon tended to be repressed by dietary sucrose (Table 18). 101 Table 15. Influence of APC gene status on expression of selected genes in small intestinal epithelial cells measured by RT-PCR. Gene name APCMin Wild-type Fold SEM P-value Change Clusterin 4.90E-03 1.20E-03 4.1 1.00E-03 0.03 LTF 1.18E-04 1.93E-05 6.1 2.20E-05 0.01 Table 16. Influence of APC gene status on expression of selected genes in colon epithelial cells measured by RT-PCR. Gene name APCMin Wild-type Fold SEM P-value Change Clusterin 1.25E-02 8.76E-04 14.3 3 .30E-03 0.04 LTF 8.93E-05 6.99E-06 12.8 2.34E-05 0.04 102 Table 17. Influence of dietary sucrose versus cornstarch on expression of selected genes in small intestinal epithelial cells measured by RT-PCR. Gene name Sucrose Cornstarch Fold SEM P-value Change IGF-II 2.78E-04 2.15E-04 1.3 5 .22E-05 0.42 IGFBP3 2.94E-05 4.80E-05 0.6 1.48E-05 0.39 PCNA 1348-02 3.1 1E-03 4.3 3 .20E-03 0.05 PIK3C2A 2.63E-03 9.5 88-04 2.7 1.02E-03 0.28 IGF] 2.53E-04 5898-05 4.3 1.34E-04 0.34 Table 18. Influence of dietary sucrose versus cornstarch on expression of selected genes in colon epithelial cells measured by RT-PCR. Gene name Sucrose Cornstarch Fold SEM P-value Change IGF-II 1.19E-03 3288-04 3.6 5.22E-05 0.03 IGFBP3 9.17E-05 5.98E-05 1.5 2888-05 0.46 PCNA 1.06E-02 6.55E-03 1.6 2.90E-03 0.36 PIK3C2A 8.8 lE-04 8.09E—04 1.1 2.29E-04 0.83 IGFl 2.42E-05 5.13E-05 0.5 1028-05 0.10 103 DISCUSSION APCMin mice have been widely used as a model for colon cancer research. Mutations in the APC gene result in production of truncated APC proteins that lack B- catenin binding domains. Thus, B-catenin is not degraded and accumulates in the cytosol. B-catenin proteins then can translocate into nucleus, form complexes with transcription factors such as T-cell factor (ch) and lymphoid enhancer factor (Lef) (Behrens et al., 1996), and subsequently increase transcription of many genes having a ch/Lef binding motif. Currently, there are more than 120 genes have been reported to be ch/Lef responsive genes (http://www.stanford.edu/~musse/pathways/targets.html). Some of these genes play an important role in colon carcinogenesis. Examples of these genes include MMP-7 (Crawford et al., 1999), Cyclin D1 (Zhang et al., 1997), C-MYC and peroxisome proliferator activated receptor 5 (He et al., 1999). We examined the NIA 15K gene library used in this experiment for genes which have ch/Lef binding motifs and have been reported as target genes in the Wnt pathway. Only some of these genes (or their homologs) were present in the NIA 15 K library, These genes included Wingless, C-MYC, cyclin D1, Axin, MYC binding protein, IGF-II, MMP-7, CD44, IRX3, and ID2. The small intestinal expression of several possible target genes in the Wnt signaling pathway was significantly different in this experiment for mice having different APC gene status (APCMin vs. wild-type). Expression of Wnt4 (homolog of Wingless) and cyclin-D2 (homolog of cyclin-D1) was significantly increased in APCMin mice compared with normal mice. It was previously reported that expression of RAMP3 (receptor activity modifying protein 3) was reduced when Wnt signaling was induced in C57MG/Wnt-l cells (Ziegler et al. 2005). In the current study, expression of 104 RAMP2 also was reduced in the APCMin mice compared with normal mice. Expression of several other genes related with the Wnt signaling pathway also were significantly altered. Expression of Dab2, an antagonist of the Wnt signaling pathway (Prunier et al. 2004; Dote et al. 2005), was down-regulated in APCMin mice. Bone morphogenetic protein receptor, type 1A (Bmprla) expression was increased in APCMin mice. BMP signaling suppresses Wnt signaling to control the duplication of intestinal stem cells, thereby preventing crypt fission and the subsequent increase in crypt number (He et al. 2004). Thus, increased expression of Bmprl a may be a compensatory mechanism to limit intestinal epithelial growth. Most experiments studying Wnt target genes have used homogenous cell populations such as colon cancer cells. In the current study, the RNA was scraped from the small intestinal epithelium of APCMin mice, which inevitably contained not only the epithelial cells but also some cells from underlying tissues (e.g. lamina propria). Furthermore, the small intestinal mucosal cell scrapings obtained from APCMin mice in this experiment would contain both tumor cells (loss both alleles of APC gene) and normal-appearing epithelial cells (with one mutated APC gene). Hence, the cell populations used for gene expression profiling in this experiment are somewhat heterogeneous. This heterogeneity of cell types may dilute our ability to detect some gene expression changes caused by APC gene mutation, and thus may reduce our ability to identify gene expression changes of some Wnt target genes in the current study. Recently, Samson et al. (2005) conditionally knocked out both alleles of the APC gene in the intestine of mice at 8-10 weeks of age using a novel inducible Ahcre transgenic line in conjunction with a loxP-flanked Apc allele. Affymetrix microarray 105 analysis was performed to compare gene expression profiles in RNA samples from the intestine of these APC knockout mice and wild-type mice. They confirmed expression changes of several previously-reported Wnt target genes, including c-Myc, CD44, Tiam l, Sema3c, Soxl7, Axin2 and Eph83 (Samson et al. 2005). However, in the Samson et al. study, both alleles ofAPC gene were deleted in all intestinal epithelial cells, which would provide for a more homogenous cell population compared with those used in our study. APCMin mice develop colonic adenomas with high frequency, and disregulation of the cell cycle is one of the hallmarks of cancer. In the present study, a large group of genes involved in cell cycle control were differentially expressed in the intestine of APCMin mice and normal mice. Expression of cyclin D2 and cyclin B 1, genes associated with increased cell proliferation, were increased in APCMin mice. D-type and B-type cyclins are essential for progression through the G1 and G2/M phases of the cell cycle. mRNA expression of clusterin was significantly increased in APCMin mice. Clusterin is implicated in immune regulation, cell adhesion, morphological transformation and cell- cell interactions. Overexpression of clusterin has been found in prostate, kidney and ovarian carcinomas. In breast tumor cells, clusterin protein accumulation correlates with the aggressiveness of breast tumor (Trougakos et al. 2004; Pucci et al. 2005). Several genes that we found to be down-regulated in APCMin mice also are involved in cell cycle control. Large tumor suppressor 2 (LatsZ) belongs to a novel tumor suppressor gene family. Recent studies reported that LatsZ negatively regulates the cell cycle by controlling Gl/S and G2/M transition (Ke et al. 2004). The decreased expression of LATSl or LATS2 mRNA was significantly associated with a large tumor size 106 (Takahashi et al. 2005). LatsZ also induces apoptosis by down-regulating anti-apoptotic proteins such as BCL-2 in human lung cancer cells (Ke et al. 2004), so decreased expression of LatsZ may lead to decreased cell apoptosis. Expression of calcium/calmodulin-dependent protein kinase ID also has been related to inhibition of apoptosis. Repression of its expression may contribute to decreased apoptosis (Y amada et al. 2005). Expression of mortality factor 4 like 1(Morf 411) was decreased in APCMin mice. Morf 411 is involved in cell growth regulation and cellular aging, and induction of this gene reverses the immortal phenotype of immortal cell lines (Bertram et al., 1999). DA82 was originally isolated as a potential tumor suppressor gene from human ovarian carcinoma (Mok et a1. 1998). Decreased expression of DA82 has been observed in several cancers, including colon cancer (Kleeff et al. 2002). Several genes involved in signal transduction were differentially expressed in APCM’" mice and normal mice in this experiment. Wnt4 was overexpressed in APCMin mice, which would further amplify B-catenin accumulation caused by APC gene mutation and contribute to increased cell proliferation and risk of intestinal tumorigenesis. Increased expression of mitogen-activated protein kinase 6 (Mapk6) in APCMin mice may also lead to increased cell proliferation. Immunoglobulin-like domain containing receptor 1 (Ildrl) was overexpressed in APCMin mice, it has been reported that expression of its cytosolic form may be related to the development and progression of cancer (Hauge et al. 2004). Expression of estrogen receptor 1 (alpha) (Esrl) was decreased in APCMin mice. Promoter hyperrnethylation of Esrl in sporadic colon adenomas has been reported (Xiong et al. 2001). This hypennethylation represses expression of Esrl and may be associated 107 with increased risk of colon cancer. TNFRSF19 is a novel member of the TNFR family that is highly expressed during embryonic development. TNFR superfamily proteins play important regulatory roles in cell proliferation, differentiation and apoptosis (Baker et a1, 1998). Decreased expression ofTNFRSF19 may contribute to decreased apoptosis in the intestinal epithelium in APCMin mice. Several genes involved in transcription regulation whose expression was influenced by APC gene status in this experiment also are associated with carcinogenesis. Overexpression of the HMGA2 gene in transgenic mice leads to the onset of pituitary adenomas (Fedele et al 2002). Expression of interferon-induced transmembrane protein 3 (Ifitm3) in the intestinal epithelium may be related with severe inflammation and increased risk of colon cancer (Hisamatsu, 1999). Transforming growth factor beta-l-induced transcript 4 (Tgfbl i4) was down- regulated in APCMin mice. Significantly decreased levels of Tgfbli4 mRNA have been reported in several types of cancers such as human brain and salivary gland tumors, and Tgfbl i4 has been suggested to have antiproliferative and tumor suppression roles (Shostak et al. 2003). Nabl is a repressor of Egrl (Russo et al 1995), and Egrl is an early growth response gene which has been implicated in cell proliferation and apoptosis. Overexpression of Egrl has a significant role in carcinogenesis and in cancer progression, especially metastasis (Kabayashi eta12002). Decreased expression ofNab 1 may result in increased expression of Egrl, thereby increasing risk of carcinogenesis. Rpal is the largest subunit of replication protein A, which is a single-stranded DNA- binding protein complex that is required for multiple processes in eukaryotic DNA metabolism, including DNA replication, DNA repair, and recombination. Mutation in 108 Rpal results in defective DNA double-strand break repair, chromosomal instability and cancer in mice, which indicates that Rpal function is essential for the maintenance of chromosomal stability and tumor suppression (Wang et al. 2005). Uxt, the ubiquitously expressed transcript, was up-regulated in APCMin mice. Although the function of Uxt is not clear, it has been previously reported that Uxt is Often overexpressed in tumor tissues (Schroer et al. 1999). Lactotransferrin (Ltf), an iron- binding protein, possesses antibacterial, antineoplastic and anti-inflammatory activity (Weinberg 2001). In experimental studies, bovine lactoferrin (bLF) significantly inhibited colon, esophagus, lung, and bladder carcinogenesis in rats when administered orally in the post-initiation stage. Furthermore, concomitant administration of bLF with carcinogens resulted in inhibition of colon carcinogenesis (Tsuda et al. 2003). However, in our study, expression of lactotransferrin in the intestinal epithelium of APCMin mice was up-regulated compared with that in normal mice. The reason for this increased expression of Ltf was not clear and needs further study. Dietary carbohydrate concentration and source have been suggested to affect risk of colon cancer. High dietary consumption of refined carbohydrates such as sucrose are associated with higher colon cancer risk (Bostick et al. 1994; Kristiansen et al. 1995; Cademi et al. 1993, 1994, 1997). Studies conducted in our laboratory indicate that consuming high-sucrose diets increases adenoma development in the small intestine and colon of APCMin mice. The second objective of the current study was to investigate the influence of dietary carbohydrate source (sucrose vs. cornstarch) on global gene expression in the intestinal epithelium of APCMin and wild-type mice using cDNA microarray profiling. Microarray analysis has been used to study gene expression 109 changes caused by dietary factors such as soy protein and vegetables in colon cancer research (van Breda et al. 2005; Xiao et al. 2005). Cui et al. (2004) used microarrays to study fructose-responsive genes in the small intestine of neonatal rats. However, the effect of dietary carbohydrate source on intestinal gene expression profiling has not been previously reported. Although small intestinal expressions of 87 genes were altered by diets containing different carbohydrate sources, we were particularly interested in genes involved in certain signaling pathways and genes directly involved in cell growth control and tumorigenesis. When the main effects of dietary carbohydrate source on gene expression in the intestinal epithelium were examined, expression of several genes involved in the insulin- like growth factor (IGF) pathway was significantly altered by dietary carbohydrate source (sucrose versus cornstarch). These genes included insulin-like growth factor 2 (IGF-II, up-regulated by sucrose versus cornstarch) and insulin-like growth factor binding protein 3 (IGFBP3, down-regulated by sucrose versus cornstarch). Expression of several other genes down-stream in the IGF signaling pathway also was influenced by dietary carbohydrate source. These genes included homolog of Grb2 (Grb7, up-regulated by sucrose) in the mitogen-activated protein kinase (MAPK) pathway and phosphatidylinositol 3-kinase (Pik302a) in the phosphatidylinositol 3-kinase (PI3K) pathway (up-regulated by sucrose). These gene expression patterns suggest that consuming high-sucrose diets is associated with elevated IGF signaling in intestinal epithelial cells. 110 Activation of the IGF Signaling pathway greatly impacts cell proliferation, differentiation, and apoptosis (Jones et al., 1995). Like IGF-I, IGF-II can bind to the IGF- I receptor, leading to receptor activation. At least two distinct signal transduction pathways have been identified for IGF-IR: one is the MAPK pathway, and increased MAPK pathway signaling leads to increased cell proliferation. Another is the PI3K pathway, wherein activation of Akt results in the phosphorylation of several other proteins that affect cell growth and cell survival. Phosphorylation of the F orkhead transcription factor (FOXO) blocks transcription of P27kip, and decreased expression of P27kip contributes to increased cell proliferation. Phosphorylation of the apoptosis- inducing protein Bad prevents cells from undergoing apoptosis. A third target of Akt is glycogen synthase kinase 3 (GSK3). Phosphorylation of GSK3 (both alpha and beta isoforms) by Akt turns off the catalytic activity. GSK3B is required for phosphorylation and degradation of B-catenin in the Wnt Signaling pathway. Thus, inactivation of GSK3B by Akt could lead to the accumulation of B-catenin, which could ultimately result in increased cell proliferation (Y u et al. 2000; F oulstone et al. 2005). Increased cell proliferation and decreased apoptosis are associated with increased risk for colon cancer. In the current study, feeding high-sucrose diets significantly induced IGF-II mRNA levels and, at the same time, reduced IGFBP3 mRNA levels in the intestinal epithelium. The ultimate outcome of these combined effects would presumably be elevated IGF signaling in the intestinal epithelium, which may contribute to increased risk of intestinal tumorigenesis in the sucrose fed animals. This result is consistent with results of other studies, in which it was reported that inducing over-expression of IGF-II in the intestine significantly increased tumorigenesis in ApcMm mice. Conversely, lll reducing IGF-II expression in the intestine resulted in reduced adenoma size and frequency (Hassan et al. 2000). It also has been observed that IGF-II mRNA levels in colon tumors are mildly elevated compared with that in normal colonic mucousa (Tricoli et al. 1986). IGF 8P3 inhibits cancer cell growth by sequestering IGF-I or IGF-II from binding IGFI-R (Cubbage et al. 1990), and can also directly inhibit tumor growth (Hong et al. 2002) and induce tumor cell apoptosis (Valentinis et al. 1995). Carcinogenesis is associated with disrupted cell cycle control that results in increased cell proliferation and growth. Among the genes whose mRNA levels were induced by the high-sucrose diet in this experiment, several function as positive cell cycle regulators. Overexpression of these genes is associated with increased cell growth and increased risk for intestinal tumorigenesis. Idb2 proteins (inhibitor of DNA binding 2) act as inhibitors of differentiation and promote cell proliferation and oncogenesis (Ruzinova et al., 2003; Lasorella et al., 2000, 2001). The Idb2 protein forms complexes with the retinoblastoma (Rb) protein and, when overexpressed, can override the Rb tumor suppressor function and lead to increased risk of oncogenesis (Lasorella et al., 2000). Proliferating cell nuclear antigen (PCNA) is an auxiliary protein (cofactor) for DNA polymerase delta and is known to be associated with S phase and DNA replication of the cell cycle (Bravo, 1986). Increased expression of PCNA is associated with increased cell proliferation, which is often related with increased risk for intestinal tumor development. Csell (chromosome segregation l-like) functions in the mitotic Spindle checkpoint, and overexpression of Csell has been reported in colonic and breast cancers (Behrens et al., 2003) 112 Among the genes whose mRNA levels were reduced by feeding high-sucrose diets in this experiment, several function as negative cell cycle regulators. Decreased expressions of these genes are associated with increased cell proliferation and increased risk of colon cancer. Tial (cytotoxic granule-associated RNA binding protein 1) is an RNA binding protein and functions as a translational silencer of cyclooxygenase-2 gene expression (Dixon et al., 2003). Overexpreesion of COX-2 is associated with increased risk of colon cancer (Eberhart et al., 1994). A null mutation for COX-2 markedly reduced the number and size of intestinal tumors in APCA716 knockout mice (Oshima et al., 1996). Fanconi anemia (FA) is an autosomal recessive disease marked by bone marrow failure, birth defects, and cancer. The FA proteins fimction at the level of chromatin during S phase to regulate and maintain genomic stability (Mi et al., 2005), so decreased expression of Fancg caused by dietary sucrose may increase genomic instability, and therefore increase risk for intestinal tumorigenesis. Sucrose suppressed the expression of Bmprla (bone morphogenetic protein receptor, type 1A). Inactivation of Bmprla in mice disturbs homeostasis of intestinal epithelial cell regeneration characterized by an expansion of the stem and progenitor cell populations, eventually leading to intestinal polyposis resembling human juvenile polyposis syndrome (He et al. 2004). It also has been suggested that 8MP signaling suppresses Wnt signaling to limit the duplication of intestinal stem cells, thereby preventing crypt fission and the subsequent increase in crypt number (He et al. 2004). Expression of several genes related to G-protein signaling was induced by high- sucrose diets. Gprc5c is a G protein-coupled receptor which can stimulate normal and aberrant cell growth and has transforming potential (Young et a1. 1986). Rapgef2 is a 113 guanine nucleotide exchange factor specific for Rapl and Rap2 (de Rooij et al. 1999) which has been implicated in a variety of cellular processes including the control of cell morphology, proliferation, and differentiation. It has been widely reported that the small GTP-binding protein Rapl has an anti-Ras and anti-mitogenic activity. However, it also has been reported that Rapl could have mitogenic effects, and is considered as a conditional oncoprotein (Daniel et al. 1998). Regulator of G-protein signaling 12 (Rgle) accelerates GTPase-activity intrinsic to the alpha subunits of heterotrimeric G- proteins and acts as a negative regulator of G-protein signaling (Dohlman et al. 1997). Although the overall effects of these alterations in expression of genes involved in G- protein signaling are not clear, their potential effects on intestinal tumorigenesis merit further investigation. Expression of phosphodiesterase 7A also was up-regulated by high-sucrose diets. cAMP-mediated effects are largely inhibitory in nature and include cell cycle arrest and apoptosis (Beavo 1995). Thus, overexpression of phosphodiesterase 7A may decrease the inhibitory effects of cAMP and increase risk for intestinal tumorigenesis. Expression of neutral sphingomyelin phosphodiesterase (Smpd3) was decreased in mice consuming sucrose. Spmd3 hydrolyzes sphingomyelin to phosphocholine and ceramide, which can inhibit cell proliferation and induce differentiation and apoptosis. (Chatterjee 1999; Stoffel et al. 2005). Decreased Spmd3 results in lower levels of ceramide, which is associated with tumorigenesis in various tissues (Duan 2005). The Aurora family kinases are pivotal to the successful execution of cell division, which together ensure the formation of a bipolar mitotic spindle, accurate segregation of chromosomes and the completion of cytokinesis. The expression of these kinases are 114 frequently deregulated in cancer and the Aur-C gene maps to 19q13.43, a region often translocated or deleted in certain cancer tissues (Crane et al. 2004). A large group of genes coding for metabolic enzymes had their expression influenced by dietary carbohydrate source in this experiment. Some genes induced by high-sucrose diets are involved in carbohydrate and fatty acid metabolism. These included phosphofructokinase (Pfld), aldolase l A (Aldoa), stearoyl-Coenzyme A desaturase 2 (Scd2), dihydrolipoamide S-acetyltransferase (Dlat), NADH dehydrogenase (ubiquinone) 1 alpha subcomplex 10 (NdufalO) and aldehyde reductase (aldose reductase)-like 6 (Aldrl6). Recent studies found that stearoyl-coenzyme A desaturase 2 also plays a role in carcinogenesis. Stearoyl-COA desaturase converts saturated fatty acids into monounsaturated fatty acids, and a positive correlation between high monounsaturated fatty acid levels and neoplastic transformation has been reported (Scaglia et a1, 2005). Endogenously synthesized monounsaturated fatty acids are involved in the control of cell proliferation, anchorage-independent growth, and survival in human transformed cells (Scaglia et a1, 2005). Phospholipid scramblase 3 (Plscr3) was down-regulated in mice consuming sucrose. Mice having a targeted deletion of PLSCR3 displayed aberrant accumulation of abdominal fat when maintained on standard rodent chow, and also developed insulin resistance, glucose intolerance, and dyslipidemia at 2 months of age (Wiedmer, 2004). Thus, the role of decreased expression of Plscr3 in the development of insulin resistance in mice fed high-sucrose diets merits further study. Defensin related cryptins (chr) and Fibronectin 1, which are involved in host defense and immunity, were found to be down-regulated in mice fed the high-sucrose 115 diet. Paneth cells in the intestinal epithelium produce several types of defensins which exert bactericidal activity. Defensin also was found to possess anticancer effects in vitro (Wong et al. 2005). Fibronectin 1 (FNl) is a homologue of fibronectin, which is a major component of the extracellular matrix and is involved in cell adhesion and migration processes. Finbronectin forms polymers which have antimetastatic effects against multiple tumor types (Pasqualini et al., 1996; Yi et al. 2001). Expressions of several genes associated with the cytoskeleton were altered by dietary carbohydrate source. Expressions of stathmin and adenylate cyclase-associated protein 1 (CAPI) were decreased by high-sucrose diets. Stathmin, which sequesters free tubulin and impedes microtubule formation, plays a pivotal role in cell motility (Currni et a1. 1999). Stathmin downregulation may be related to defective cell migration in the intestinal crypt. Cyclase-associated proteins are highly-conserved actin monomer binding proteins and depletion of CAPl can result in cytoskeletal defects (Bertling et al. 2004) The establishment and maintenance of adhesion at adherens junctions requires the stabilization of cell adhesion molecules at specific sites of cell—cell contact. Cell adhesion molecules, in particular E- and N-cadherin, engage in homophilic interactions and thereby promote the recruitment of or-catenin by B-catenin, which in turn directly or indirectly binds actin filaments. Actin filaments are believed to stabilize E-cadherin—B- catenin—or-catenin complexes (Lecuit 2005). Thus, increased expression of or-catenin in mice fed the high-sucrose diet may have some consequence on stability of adherens junctions. 116 mRNA expression of several genes in the proximal small intestinal epithelium which were found to be significantly influenced by dietary carbohydrate source or APC gene status using cDNA microarray analysis was further assessed using RT-PCR analysis. Expression of PCNA was significantly (P = 0.05) increased by high dietary sucrose, and feeding diets containing high-sucrose also tended to alter expression of IGF- II, IGFBP3 and PIK3C2A in the same direction as identified in cDNA microarray analysis. Expression of clusterin and lactotransferrin were confirmed to be significantly up-regulated in APCMin mice measured by RT-PCR. Overall, the results from RT-PCR analysis are support the findings of cDNA microarray analysis. Expression of several genes in colonic epithelial cells obtained from the same mice was measured using RT-PCR. Expression of IGF-II in the colonic epithelium was significantly (P < 0.05) up-regulated in mice consuming high-sucrose diets. Overexpression of IGF-II in colon has been associated with increased risk for colon Min cancer. In Apc mice, it was found that reducing IGF-II expression in the intestine by Mm” mice with IGF-II gene knockout mice resulted in reduced adenoma size crossing Apc and frequency. Conversely, inducing over-expression of IGF-II in the intestine significantly increased tumorigenesis in ApcMW+ mice (Hassan et al. 2000). The IGF-II gene is maternally imprinted in that it is expressed only from the paternal copy of the gene. Loss of imprinting (LOI) of the insulin-like growth factor II gene is an epigenetic alteration that results in a modest increase in IGF-II expression, and it is present in the normal colonic mucosa of about 30% of patients with colorectal cancer (Sakatani et al. (2005) 117 In summary, cDNA microarray analysis identified 109 genes whose expression in small intestine was significantly different in APCMin mice and wild-type mice. Many of these genes are associated with cell growth control and tumorigenesis (increased expressions of Clu, Ccnd2, Ccnbl, Btg4, Anxal, Gsptl; decreased expressions of Camkld, septin 2, LatsZ, Dab2, Morf4ll). These observations indicate that APC gene mutations have a broad influence on expression of many genes associated with intestinal tumorigenesis. Consumption of high-sucrose diets significantly altered expression of 87 genes in the small intestinal epithelium when compared to consumption of high- comstarch diets. High-sucrose diets also increased expression of IGF-II and decreased expression of IGFBP3. Collectively, these observations suggest that increased risk of colon carcinogenesis caused by high levels of dietary sucrose is associated with elevated IGF signaling in the intestinal epithelium. 118 CHAPTER IV. INFLUENCE OF DIETARY CARBOHYDRATE (SUCROSE VERSUS CORN STARCH) AND PROTEIN (CASEIN VERSUS SOY) SOURCES ON INTESTINAL TUMORIGENESIS IN SULINDAC-TREATED APCMIN MICE 119 ABSTRACT Our previous research indicated that dietary sucrose increased adenoma numbers in the proximal small intestine and cell proliferation in colon crypts in APCMin mice after 10-weeks of dietary treatment. Soy protein has been demonstrated to inhibit colon cancer development in carcinogen-induced colon cancer models. In the current study, the non- steroidal anti-inflammatory drug sulindac was added to all diets at 100 mg/kg to retard small intestinal tumorigenesis of APCMin mice, thereby allowing them to survive for a longer period of time before experiencing significant morbidity secondary to small intestinal adenoma development. The objective of this study was to utilize this extended- progression protocol to assess the impact of dietary carbohydrate and protein sources on colonic adenoma development and progression in APCMin mice. Mice were randomly assigned at four weeks of age to one of four modified AIN- 93G diets containing different protein (soy versus casein) and carbohydrate (cornstarch versus sucrose) sources. Mice were fed ad libitum for 16 weeks or until they became moribund. Weekly body weights, intestinal adenoma development and colon crypt epithelial cell kinetics were determined. Mice consuming either sucrose or soy flour gained significantly more weight than those consuming cornstarch (P < 0.01) or casein (P < 0.05), respectively. Feeding sucrose promoted colon tumor development in APCMin mice compared with feeding cornstarch, with greater tumor incidence (59 vs. 30%, P < 0.01), greater tumor numbers (0.94 vs. 0.46 per mouse P < 0.01) and heavier tumor burden (6.8 vs. 2.5 mm3 per mouse, P < 0.01). Ki67 antigen immunohistochemistry indicated that mice fed sucrose had higher rates of epithelial cell proliferation in colon crypts compared to mice fed cornstarch (labeling index: 40 versus 32% for sucrose and cornstarch, 120 respectively, P < 0.01; proliferative zone: 61% versus 53%, P < 0.01), and reduced apoptosis (by TUNEL assay, apoptotic index: 3.8 versus 5.6%, P < 0.05). Feeding diets containing soy flour promoted small intestinal adenoma development in APCMin mice compared with feeding casein, with increased sizes of adenomas (1.37 versus 1.04 mmz, P < 0.01) and greater tumor burden (64 versus 42 mm2 per mouse, P < 0.01). Dietary soy protein source had no effects on colon tumor development. Seven (7/35; 20% incidence) female mice consuming soy flour developed mammary gland tumors, whereas none (0/26; 0% incidence) of the female mice consuming casein-based diets developed mammary tumors (P < 0.05). In summary, diets containing high sucrose concentrations significantly increased colonic tumor incidence compared with cornstarch in APCMin mice. Dietary soy protein source had no effects on colon tumor development. Female APCMin mice consuming soy flour had significantly higher incidence of mammary gland tumors compared with that consuming casein-based diets. 121 INTRODUCTION Our previous research indicated that high-sucrose diets increased proximal small intestinal adenoma numbers and colonic epithelial cell proliferation in APCMmmice. However, high-sucrose diets did not significantly influence colonic adenoma development. We speculate that the lack of effect of sucrose on colonic adenoma development in the previous experiment (Chapter 11) may be due to the relatively short time period (10 weeks) of dietary treatment for APCMin mice, which may not have been sufficient in duration for these diets to exert their full impact on colon tumor progression. APCMin mice typically exhibit morbidity at 14-16 weeks of age due to rapid adenoma development in the small intestine, which causes bleeding, severe anemia and weight loss. Colon tumors in APCMin mice typically develop more slowly than small intestinal adenomas. Sulindac is a non-steroidal anti-inflammatory drug which has been shown to reduce the rate of small intestinal tumor development in APCMin mice. However, sulindac generally does not reduce colonic tumor growth in studies using APCMin mice (Jacoby et al., 1996; Chiu et al., 1997; Sorensen et al., 1998). By adding a low dose of sulindac to the diet (100 mg/kg), we can expand the life-span of APCMin mice by inhibiting small intestinal tumor development. This allows more time for development of the colonic tumors, therefore increasing the possibility to detect dietary effects on colonic tumor development. The objective of this study was to utilize this extended-progression protocol to assess the impact of dietary carbohydrate and protein sources on colonic adenoma development and progression in APCMm mice. As was the case in the previous 122 experiment, the carbohydrate sources examined were sucrose and cornstarch. We also were particularly interested in assessing the impact of dietary protein source on intestinal adenoma development in APCMin mice, as soy protein has been demonstrated to be protective against development of colon cancer in rodent models of human colon cancer. To date, the effects of soy protein feeding have not been fully explored in APCMin mice. Hence, we also compared the impact of dietary protein source (soy versus casein) on intestinal adenoma development in APCMin mice. 123 MATERIALS AND METHODS Animals and Diets: . Mice for the study were produced by mating C5 7BL/6J male APCMin mice with normal C57BL/6J female mice. The resulting offspring (50% normal and 50% APCMin based on simple Mendelian inheritance) were randomly assigned at weaning (4 weeks of age) to one of four diets containing different protein and carbohydrate sources. Each group had similar numbers of males and females. Sixty mice were assigned to group. Of these, we expected 50% to be APCMin mice, thus we expected appoximately 30 APCMin mice per treatment group. Mice used in this study were genotyped by an allele-specific PCR procedure to identify APCMin progeny (Jacoby et al., 1996). Mice were housed in a room with constant temperature and humidity (23°C, 60% humidity) and a 12: 12 hour light-dark cycle. Animal care and feeding were conducted with approval of the Michigan State University All-University Committee on Animal Use and Care. The treatment design was a 2 X 2 factorial having two carbohydrate sources (cornstarch or sucrose) and two protein sources (casein or soy flour). The diets (Table 19) were based on modified AIN-93G diets (Reeves et al., 1993). Additionally, all diets contained 100 mg/kg sulindac. Mice were fed diets ad libitum for the duration of the study (Sixteen weeks or until individual animals exhibited body weight loss of more than 10%). 124 Table 19. Composition of diets for experiment 3 (g/kg diet). Ingredient Casein & Casein & Soy & Soy & Starch Sucrose Starch Sucrose Defatted Soy Flour 0.00 0.00 411.36 411.36 Casein 222.20 222.20 0.00 0.00 Corn Starch 504.19 0.00 382.41 0.00 Sucrose 0.00 504.19 0.00 382.41 Soybean Oil 150.00 150.00 150.00 150.00 Cellulose 67.38 67 .38 0.00 0.00 AIN-93G-MX 38.90 38.90 38.90 38.90 AIN-93-VX 11.10 11.10 11.10 11.10 L-cysteine 3.30 3.30 3.30 3.30 Tert-Butylhydroquinone 0.03 0.03 0.03 0.03 Choline Bitartrate 2.80 2.80 2.80 2.80 Sulindac 0.10 0.10 0.10 0.10 125 Measurements: After sixteen weeks of dietary treatment, mice began to exhibit morbidity due to intestinal tumor development. At this time, all mice were euthanized by C02 asphyxiation. Liver samples were collected from each mouse to determine the APC gene status using PCR (Jacoby et al., 1996). The entire intestine was removed, opened longitudinally, rinsed with water, pinned on cardboard, and fixed with 10% neutral buffered formalin overnight. The numbers and sizes of adenomas in each section were determined with the aid of a stereo-microscope as described in Chapter II. A one- centimeter section was removed from the distal colon and processed for immunohistochemistry analyses of epithelial cell kinetics (Ki67, PCNA, TUNEL assay) in colonic crypts as described in Chapter II. We also noted that several female mice developed mammary tumors in this experiment. When detected, these mammary tumors were excised, weighed, and fixed in neutral buffered formalin to allow for histological grading at a later date. Statistical Analyses: Data were analyzed using the General Linear Models procedure of SAS (Version 8.2). The experimental design was a completely randomized design with a 2 X 2 factorial arrangement of treatments (protein source, carbohydrate source). When significant treatment effects or interactions were detected (F-test significant at P < 0.05), the Least Significant Difference method was used to compare appropriate main effect or treatment means. The incidence rates of colonic adenomas and mammary tumors in mice were analyzed using the Frequency procedure of SAS. 126 RESULTS Body weight: Weekly body weights of APCMin mice consuming the four diet combinations are shown in Figure 13. APCMin mice fed diets containing soy flour gained more weight during the study than mice fed diets containing casein (14.1 i 0.28 vs. 13.2 i 0.32 grams; P < 0.05). Mice fed diets containing sucrose also gained more weight than mice fed diets containing cornstarch (14.5 i 0.31 vs. 12.8 i 0.29; P <0.01). Dietary intakes by mice were not measured in the study. Effects of diets on intestinal tumorigenesis: There were no interactions between effects of carbohydrate and protein sources on intestinal tumorigenesis, so the main effects of carbohydrate and protein sources on intestinal adenomas are presented. When adenoma development in the whole small intestine was compared, there were no significant differences in adenoma numbers, sizes or total burden for APCMin mice fed diets containing cornstarch or sucrose (Table 20). Mice that consumed soy as the primary protein source had small intestinal adenomas that were significantly larger in size when compared to those observed in mice consuming casein, which resulted in the soy-fed mice having a greater total burden (number X size) of small intestinal adenomas when compared to casein-fed mice (Table 20). Dietary carbohydrate source significantly influenced the numbers and average sizes of adenomas in proximal small intestine (Figure 14, Figure 15). These effects were similar to that observed in the first experiment (Chapter 11) despite adding sulindac in the diets of the current study. APCMin mice fed sucrose-based diets had Significantly greater numbers of adenomas in the proximal third of the small intestine than mice fed comstarch-containing diets (18.1 vs. 13.3, P < 0.05; Figure 14). However, the average 127 size of adenomas in the proximal small intestine was greater in mice fed cornstarch- containing diets (1.96 vs. 1.26 mmz, P < 0.05; Figure 15). Overall, the total adenoma burden in the proximal small intestine for the two groups was not significantly different (Figure 16). Mice that consumed soy as the primary protein source had a significantly greater number of adenomas in the distal small intestine compared to mice that consumed casein. (17.9 vs. 13.6, P < 0.05; Figure 17). Adenomas in the medial and distal small intestine of mice consuming soy also were significantly larger in average size when compared to those in mice consuming casein (Figure 18). The total tumor burdens in the medial and distal regions of the small intestine of APCMin mice consuming soy protein also were significantly greater than those in mice consuming casein (Figure 19). Colonic adenoma incidence, numbers, and burdens are presented in Table 21. The incidence of colonic adenomas in APCMin mice in this experiment was significantly greater in mice consuming sucrose-based diets (58.7%) when compared to mice consuming comstarch-based diets (30.1%). Averaged across all APCMin mice, those consuming sucrose had significantly greater colonic adenoma numbers per mouse, larger average size of colonic adenomas, and greater total adenoma burden per mouse relative to that in mice consuming cornstarch (Table 21). Among colonic tumor-bearing mice only, there was no difference in colonic adenoma number per mouse. Thus, all of the differences observed among treatments in adenoma numbers were explained by the difference in adenoma incidence among APCMin mice. Dietary protein source did not influence colonic adenoma incidence. 128 Although female APCMin mice are known to be susceptible to mammary tumorigenesis, no research conducted to date has indicated that diet can influence mammary tumor development in this model. Mammary tumor incidence of female APCMin mice is presented in Table 22. In the current study, seven (7/35; 20% incidence) female mice consuming soy flour developed mammary gland tumors, whereas none (0/26; 0% incidence) of the female mice consuming casein-based diets developed mammary tumors. This difference in mammary tumor incidence was statistically significant (P < 0.05). Dietary carbohydrate source did not influence mammary tumor incidence. 129 3° ‘ —A— Soy-Sucrose —O—Casein-Sucrose —*—Soy-Comstarch : 26 - +Casein-Cornstarch rim-0"“ f—F" 28- BodyWeight (grams) 10 I I I I I I I I I I I T I fir I r I 012 3 4 5 6 7 8 910111213141516 Weeks of Dietary Treatment Figure 13. Body weights of APCMin mice consuming diets containing different protein and carbohydrate sources. 130 Table 20. Small intestinal adenoma numbers, average Sizes of small intestinal adenomas, and total burden of small intestinal adenomas in APCMm mice consuming diets based on differing protein and carbohydrate sources (main effects; means i standard errors). Carbohydrate Source Protein Source Parameter Cornstarch Sucrose Casein Soy (n=63) @54) (n=50) (n=67) Adenoma number 43.5 :t 3.3 46.4 i 3.5 43.1 is 3.7 46.7 i 3.1 Average size (mmz) 1.27 i 0.06 1.13 d: 0.07 1.04 :l: 0.07a 1.37 d: 0.06b Total burden (mm2) 54.5 :I: 4.5 52.0 :1: 4.8 42.4 :t 5.021 64.2 i 4.3b a’bProtein source effect (P < 005): 131 30- El Sucrose-based diet E1 Cornstarch-based diet 25- ‘- a g 20- E c 15‘ E o 10- 5 'U 5- < Proximal Small Medial Small Distal Small Intestine Intestine Intestine Figure 14. Main effect of dietary carbohydrate source on small intestinal adenoma numbers in APC“. * All APCMin mice had small intestinal adenomas. a’b Significant diet effect (P<0.01). 132 25. Sucrose-based diet to" b Cornstarch-based diet E 2- § § 1.5- a (I) re E 1- o c 8 < 0.5" Proximal Small Medial Small Distal Small Intestine Intestine Intestine Figure 15. Main effect of dietary carbohydrate source on small intestinal average adenoma sizes in APC M'" a,b Significant diet effect (P<0.05). 133 35 - El Sucrose-based diet :5" El Cornstarch-based diet E 30- E E, 25‘ “g 20- 3 ED 15. E o 10- 5 13 5‘ < n :1 , . . , u . f A Proximal Small Medial Small Distal Small Intestine Intestine Intestine Figure 16. Main effect of dietary carbohydrate source on total small intestinal adenoma burden in APCMm mice 134 30_ El Soy-based diet [I Casein-based diet 25- I- 3 20- a E c 15- E o 10- 5 'c 5- < Proximal Small Medial Small Distal Small Intestine Intestine Intestine Figure 17. Main effect of dietary protein source on small intestinal adenoma numbers in APCMm mice. a,b Significant diet effect (P<0.05). 135 2,5- El Soy-based diet D Casein-based diet 2- :5" E 1.5- a I .5 1- U) E o 0.5- c e 'u < c . . * Proximal Small Medial Small Distal Small Intestine Intestine Intestine Figure 18. Main effect of dietary protein source on average small intestinal adenoma sizes in APCM‘" mice. a,b Significant dict effect (P<0.05). 136 4o . El Soy-based diet 35- CICasein-based diet .1‘ 30- E 25- : 20- a o '2 , b 5 15 g 10 2 5‘ a, 0 . 2 Proximal Small Medial Small Intestine Intestine Intestine Figure 19. Main effect of dietary protein source on total small intestinal adenoma burden in APCM‘". a,b Significant diet effect (P<0.05). 137 Table 21. Colonic adenoma numbers, average sizes of colonic adenomas, and total burden of colonic adenomas in APCMm mice consuming diets with protein and carbohydrate sources (main effects; means :t standard errors). Carbohydrate Source Protein Source Sucrose Parameter (n=54) A11 APCMill mice: n e a Adenoma Incidence (%) 58.7 a Adenoma number 0.94 i 0.12 Adenoma size (mm3) 4.95 i 0.8421 Adenoma burden (mm3) 6.83 :t 1.1721 Tumor-bearing mice: Adenoma number 1.57 d: 0.15 Adenoma size (mm3) 8.45 :t 1.49 Adenoma burden (mm3) 11.23 :t 2.05 Starch (n=63) 30.1b 0.46 3: 0.11b 1.32 3: 0.77b b 2.51 :1: 1.08 1.57 :1: 0.19 4.40 i 1.96 8.26 i 2.69 Soy (n=67) 44.1 0.70 i 0.11 4.02 :1: 0.74 5.84 i 1.04 1.61 i016 8.101 1.63 12.08 i 2.24 Casein (n=50) 44.7 0.70 :1: 0.13 2.25 :t 0.87 3.50 :1: 1.21 1.53 :t 0.18 4.75 :t 1.85 7.40 :t 2.53 a’bCarbohydrate source effect (P <0'05)° 138 Table 22. Mammary tumor incidence in female APCMin mice consuming diets based on differing protein and carbohydrate sources. Carbohydrate Source Protein Source Parameter Cornstarch Sucrose Casein Soy @=36) (n=25) (n=26) (n=3 5) Mammary tumor a b incidence (%) 11 (4/36) 12 (3/25) 0 (0/26) 20 (7/35) a’bProtein source effect (P < 0.05). 139 Effects of diets on intestinal crypt cell kinetics: Ki-67 antigen was used as a surrogate marker for cell proliferation in this study. Mice consuming sucrose had Significantly greater total crypt Ki-67 labeling index (labeling index = labeled cells per hemicrypt / crypt height) when compared to mice consuming cornstarch (Table 23). Dietary sucrose also significantly increased Ki-67 proliferative zone (proliferative zone = position of highest labeled cell in crypt / crypt height) relative to cornstarch, indicating that sucrose feeding expanded the overall zone of cell proliferation in the colon. These results were further confirmed by our observation that Ki-67 labeling index was increased in the middle and top (luminal) thirds of the colonic epithelium when mice consumed sucrose versus corn starch. Dietary protein source did not influence Ki-67 labeling indices in this experiment. Proliferating cell nuclear antigen (PCNA) is expressed in colonic epithelial cells that have not undergone terminal differentiation. Total crypt PCNA labeling index and proliferative zone were not influence by diet in this experiment (Table 24). However, PCNA labeling in the upper one-third of colonic crypts was significantly greater in mice consuming sucrose when compared to mice consuming cornstarch. This observation indicates that there were a slight upward shift in PCNA labeling in mice consuming sucrose, and would suggest a delay in terminal differentiation as colonic epithelial cells migrate up the crypt axis when mice consumed the high-sucrose diet. Dietary protein source did not influence PCNA labeling indices in this experiment. As colonic epithelial cells migrate to the crypt surface, they undergo programmed cell death and are eliminated from the crypt apex. Programmed cell death of colonic 140 epithelial cells was assessed by the TUNEL assay, and these results are presented in Table 25. Mice consuming the high-sucrose diets had significantly fewer TUNEL- positive cells in the total crypt and the upper third of the crypts when compared to mice consuming the comstarch-based diets. This suggests that colonic epithelial cells were less likely to undergo programmed cell death at the crypt surface when mice consumed the high-sucrose diets. Mice consuming the soy diets had significantly fewer TUNEL- positive cells in the total crypt and the middle third of the crypts when compared to mice consuming the casein-based diets. 141 Table 23. Mean heights of colonic crypts and Ki-67 antigen expression in colon of APCMm mice consuming diets based on differing protein and carbohydrate sources (main effects; means i standard errors). Carbohydrate Source Protein Source Parameter Sucrose Corn Starch Soy Casein (n=22L Jn=18) (n=22) (n=18) Crypt Height (cells) 22.1 e 0.4 21.3 e 0.5 21.9 e 0.4 21.5 3: 0.5 Total L1 0401i00123 0320i0013b 0.366i0.012 036020.013 Bottom 1/3 LI 0.643 i 0.025 0.601 :1: 0.028 0.603 i 0.025 0.641 3: 0.028 Middle 1/3 LI 0 485 i 0 024a 0 350 i 0 026b 0.447 i 0.024 0.388 :1: 0.026 Top 1/3L1 007910014“ 0022,5001; 0.049e0.014 00522.0.015 a,b Carbohydrate source effect (P < 0.05). 142 Table 24. PCNA expression in colon of APCMin mice consuming diets based on differing protein and carbohydrate sources (main effects; means :1: standard errors). Carbohydrate Source Carbohydrate Source Parameter Sucrose Corn Starch Soy Casein (n=32) (n=3 8) (n=40L (n=30) Total LI 0.591 i 0.019 0.557 d: 0.017 0.589 :t 0.016 0.559 :t 0.019 Proliferative 0.708 t 0.019 0.660 :1: 0.017 0.697 :1: 0.017 0.671 :1: 0.020 Zone Bottom 1/3 LI 0.818 i 0.018 0.844 :1: 0.016 0.831 :1: 0.016 0.831 t 0.018 Middle 1/3 LI 0.756 t 0.032 0.71221: 0.028 0.775 :t 0.028 0.693 :t 0.033 Top 1/3 L1 0198 i 0.02321 0.114 3. 0.020 b 0.159 i 0.020 0.156 i 0.023 a’bCarbohydrate source effect (P < 0.05). 143 Table 25. TUNEL assay results in colon of APCMin mice consuming diets based on differing protein and carbohydrate sources (main effects; means i standard errors). Carbohydrate Source Sucrose Corn Starch (n=2 1) (n=23) 0.038 i 0.0063 Parameter Total LI Middle 1/3 LI 0.00] i 0.000 0.000 i 0.000 TOP “3 H 0.111 e 0.019a 0.056 3: 0.006b Bottom “3 L1 0.001 :t 0.000 a 0.0000: 0.000b 0.167:i_-0.018b Protein Source Soy Casein (n=22) (n=22) 0.039 i 0.006c 0.055 i 0.006 d 0.001 i 0.000 0.001 :t 0.000 d 0.002 :t 0.000cl 0.163:1:0.018f 0.000 3. 0.000 0.115 e 0.018e a’bCarbohydrate source effect (P < 0.05). c’dProtein source effect (P < 0.05). e’fProtein source effect (P < 0.06). 144 DISCUSSION APC gene mutations occur in the early stage of colonic carcinogenesis (Fearon et al., 1990; Kinzler et al., 1996). Therefore, APCMin mice represent an excellent model to study the influence of dietary factors on colon cancer progression. However, a major disadvantage of using APCMin mice for colon cancer research is that most tumors typically are located in the small intestine. Because the small intestinal adenomas in APCMin mice grow rapidly and cause bleeding, severe anemia and weight loss, APCMin mice typically display signs of morbidity by 14-16 weeks of age. We previously examined the effects of high-sucrose diets on colon tumorigenesis in APCMin mice (Chapter 11). Due to morbidity associated with small intestinal adenomas, dietary treatments had to be ended after 10 weeks. We found that high- sucrose diets significantly increased adenoma numbers in the proximal small intestine. In the previous experiment, adenoma development in the colon was not significantly influenced by dietary carbohydrate source. However, high-sucrose diets significantly increased the overall rate of cell proliferation in the colonic crypts of mice consuming sucrose compared to that in mice consuming cornstarch in the previous study. Because colon tumors develop more slowly than small intestinal tumors, we speculated that a longer duration of dietary treatment would assist in identifying carbohydrate source effects on colon tumor development in this model. Sulindac has been widely studied as potential chemopreventive agent for colon cancer. Previous studies in APCMin mice have shown that sulindac significantly reduces the number and size of small intestinal adenomas, but does not significantly affect colon tumor development in most studies (Chiu et al., 1997; Sorensen 1998). In the current 145 study, a low dosage of sulindac (100 mg/kg diet) was added to each of the diets to limit the development of small intestinal adenomas. This approach significantly extended the duration of diet treatments from 10 weeks to 16 weeks in this Study. Feeding sucrose is associated with greater proliferation in colonic crypts, as well as a greater number of large aberrant crypt foci (Cademi et al., 1993, 1997; Kristiansen et al., 1995; Poulsen et al., 2001). Cademi et al. (1994) reported that rats fed high-sucrose diets had significantly greater numbers of colonic adenomas per rat compared with those fed cornstarch-based diets in a DMH-induced rat colon cancer model. The adenocarcinomas in the rats fed sucrose were larger and had greater invasive potential than those in rats fed the comstarch-based diets, although the incidence of total intestinal tumors was not affected by the different carbohydrate sources (Cademi et al., 1994). In this study, we demonstrated that feeding high sucrose diets significantly increased colon adenoma incidence compared with feeding cornstarch diets in APCMin mice. We believe this is the first study to directly demonstrate that high-sucrose diets stimulate colon tumor development. The colonic epithelium is in a constant state of cell renewal. Abnormal net increase of cell population in the colonic crypts is associated with increased risk of colon cancer (Lipkin et al., 1974. 1984; Bleiberg et al., 1985; Terpstra et al., 1987; Ponz de Leon et al., 1988; Mills et al., 2001). Increased cell population in colonic crypts would result from increased cell proliferation, delayed cell differentiation, or reduced programmed cell death of colonic epithelial cells. In the current study, Ki-67 antigen was used as a surrogate marker for cell proliferation. PCNA is expressed in colonic epithelial cells that have not undergone 146 terminal differentiation and was used in this study to assess cell differentiation. The TUNEL assay was used to identify cells undergoing programmed cell death. Immunohistochemistry analyses indicate that feeding high-sucrose diets was associated with increased cell proliferation, delayed cell differentiation, and reduced programmed cell death of colonic epithelial cells. These observations are consistent with the observed increase in colonic adenoma incidence when mice consumed the sucrose-based diets. Although it has been demonstrated that consumption of soy flour protects against colon cancer in carcinogen-induced rat colon cancer models (Bennink et al., 1999, 2000), consumption of soy flour did not have any protective effects on colon tumor development in the current study. Rather, soy consumption promoted tumorigenesis in the medial and distal small intestine. The mechanisms whereby dietary soy might stimulate colonic adenoma development are unclear. Only one study to date has tested the effects of soy protein isolates containing different levels of isoflavones in APCMin mice (Sorensen et al., 1998), and it found that soy isoflavones did not influence the development of intestinal adenomas. In the current study, TUNEL analysis indicated that the apoptotic rates of colonic epithelial cells of mice consuming soy-based diets were significantly reduced compared to that observed in mice consuming casein-based diets. However, the colonic epithelial cell proliferation rate was not different for mice consuming soy or casein. We were unable to assess cell kinetics in small intestine in the current study due to difficulties in preparing intact, full-length small intestinal crypts. Future studies on this may provide some insights into the mechanisms whereby soy diets promote small intestinal adenoma development in APCMin mice. 147 Female APCMin mice in 57BL/6J background occasionally develop mammary gland tumors (Moser et al., 1992, 1993), which indicate a tumor suppressor role for APC within the mammary epithelium. However, the mammary gland tumor incidence observed in APCMm mice is low because APCMin mice die around 15-16 weeks of age due to rapid small intestinal tumorigenesis. In the current study, female APCMin mice consuming soy flour had significantly greater incidence of mammary gland tumors compared those consuming casein-based diets (20% vs. 0%, P < 0.05) after 16 weeks of dietary treatment. No research conducted to date has reported that diet can influence mammary tumor development in this model. The influence of soy protein on mammary gland tumor development has been extensively studied using carcinogen-induced animal models. The isoflavone genistein has been considered to be responsible for the effects of soy protein on breast cancer development (Messina et al., 2001). The effects of dietary soy products on mammary tumor development are still controversial. Early exposures in life to high concentrations of genistein were found to be protective against DMBA-induced mammary tumor development (Lamartiniere et al., 1995; Murrill et al., 1996). This effect is believed to results from the stimulation by genistein of early mammary gland maturation, making these animals less susceptible to mammary gland tumor development when exposed to carcinogens later in life (Messina et al., 2001). Hawrylewicz et al. (1995) reported that, after induced breast tumors by injecting MNU, rats consuming soy proteins developed significantly fewer tumors, and tumors in the soy-fed rats were less aggressive than those observed in casein-fed rats. However, in ovariectomized athymic mice, dietary soy protein or genistein have been demonstrated to stimulate the growth of subcutaneously 148 implanted MCF-7 cells (estrogen-dependent human breast cancer cells) in a dose- dependent manner (Hsieh et al., 1998; Allred et al., 2001). Effects of isoflavones on mammary tumor development also were studied using MMTV-neu mice, which spontaneously develop mammary tumors due to overexpression of the Erb8-2/neu/HER2 oncogene (J in etal. 2002). Feeding MMTV-neu mice with AIN-93G diets containing isoflavones from 7 weeks of age significantly delayed mammary tumorigenesis compared to the feeding of a low-isoflavone control diet. However, feeding dietary isoflavones did not reduce the numbers or sizes of tumors in these mice once tumors formed (J in et al., 2002). In the current study, feeding APCMin mice soy flour from 4 weeks of age significantly increased mammary gland tumor incidence compared to feeding mice with casein-based diets. Unlike ovariectomized athymic mice, female APCMin mice have normal ovary function and estrogen production. Despite having a normal estrogen background in the mice used in our study, soy protein containing isoflavones still was able to stimulate mammary tumor development. Further studies on the effects of soy- containing diets on mammary tumorigenesis in APCMin mice are warranted. In summary, this experiment demonstrated that feeding high-sucrose diets significantly increased colonic tumor incidence compared with feeding comstarch-based diets in sulindac-treated APCMin mice. High-sucrose diets also caused changes in epithelial cell kinetics in the colonic crypts (increased proliferation, delayed differentiation and reduced programmed cell death) consistent with increased risk of colon cancer. Previous studies in rodents indicate that high-sucrose intakes cause compensatory hyperinsulinemia, which leads to increased expression of IGF-I in liver 149 (Chapter 11; Jones et al., 1995; Giovannucci 1995). A previous study in our laboratory also suggested that high dietary sucrose induces local overexpression of IGF-II in colon (Chapter III). Elevated signaling via the IGF pathway is related to accelerated cell cycle and repressed apoptosis, which ultimately leads to increased risk for colon cancer development (Yu et al., 2000). Further research is warranted to determine if the stimulatory effects of high-sucrose diets on colonic tumorigenesis are associated with increased IGF signaling on colonic epithelial cells, or if other mechanisms explain this effect. 150 CHAPTER V. OVERALL SUMMARY AND CONCLUSIONS 151 Colon cancer is the second most common cause of cancer mortality and claims more than 50,000 lives every year in the United States (ACS, 2004). Dietary factors strongly modify colorectal cancer risk and approximately 66% to 75% colon cancer may be prevented by adequate diets and physical activity (AICR/WCRF, 1997). Diets rich in refined carbohydrates such as sucrose have been associated with high colon cancer risk (Tuyns et al., 1988; La Vecchia et al., 1993; Bostick et al., 1994;Centonze et al., 1994; De Stefani et al., 1998). Experiments using carcinogen-induced colon cancer also have shown that feeding rats with high sucrose diets promoted growth of aberrant crypt foci, a proposed precancerous lesion of the colon, and also were associated with increased proliferation in colon crypts compared with feeding cornstarch diets (Kristiansen et al., 1995; Cademi et al., 1993, 1997; Poulsen et al., 2001). Colon adenocarcinomas in carcinogen-treated rats consuming high-sucrose diets were significantly larger and had more invasive potential compared with those in rats consuming cornstarch (Cademi et al., 1994). However, to date high dietary sucrose has not been demonstrated to increase colon cancer incidence in animal studies. Furthermore, the underlying mechanisms whereby high dietary sucrose may increase colon cancer risk are unclear. APCMin (multiple intestinal neoplasia) mice carry a germline mutation in the adenomatous polyposis coli (APC) gene (Su et al., 1992), which is similar to APC gene mutations observed in humans with F AP (familial adenomatous polyposis). FAP patients develop hundreds of colonic adenomas and are at increased risk of colon cancer (N ishisho et al., 1991). APC gene mutations also are commonly present in sporadic colon cancers (Powell et al., 1992). Kinzler and Vogelstein (1996) have proposed a “gatekeeper” function for APC gene in colorectal epithelia in which APC is responsible 152 for maintaining a constant census in renewing cell populations. Therefore, APCMin mice represent a useful model to study diet and gene interactions during colon carcinogenesis. Animal studies suggest that high intakes of sucrose induce compensatory hyperinsulinemia by causing a decline in insulin sensitivity in the liver and peripheral tissues. Hyperinsulinemia may increase IGF-I (insulin-like grth factor 1) production in liver and free IGF-I in the circulation (Giovannucci, 2001). Evidence also indicates possible associations between altered gene expressions of IGFBP3 (insulin-like growth factor binding protein 3) and IGF-II (insulin-like growth factor II) in the colon epithelia and colon carcinogenesis (Hassan et al., 2000; Kirman et al., 2004). The purpose of the current research was to determine if diets high in sucrose (versus cornstarch) promote colon carcinogenesis in APCMin mice, and if this action was associated with elevated IGF signaling and increased cell proliferation in colonic epithelia. To confirm the hypothesis, three experiements were conducted. The objectives of the first study were to test the effects of diets containing sucrose or cornstarch as the sole carbohydrate source on intestinal adenoma development in APCMin mice, and possible associations between elevated blood IGF concentrations caused by high dietary sucrose and increased colon cancer risk. APCMin mice (n = 48) were randomly assigned at 4 weeks of age to one of two modified AIN-93G diets containing either sucrose or cornstarch (523 g/kg of diet) as the sole carbohydrate source. Diets were fed ad libitum for 10 weeks. Weekly body weights, intestinal adenoma development and cell kinetics (Ki67, PCNA and TUNEL labeling) in colonic crypts were measured. The levels of glucose and insulin in the circulation, as well as mRNA expressions of IGF-I, IGF-II, IGF BF] and IGFBP3 in liver also were determined. 153 In comparison to cornstarch, sucrose feeding promoted weight gain in mice (P < 0.05). APCMin mice fed sucrose had significantly greater numbers of adenomas in the proximal third of the small intestine than mice fed cornstarch (21.8 vs. 13.1, P < 0.01). However, the average size of proximal small intestinal adenomas was larger in comstarch- versus sucrose-fed mice (2.1 vs. 1.1 mmz, P < 0.01). The total number of adenomas present in the entire small intestine tended to be greater in sucrose-fed mice (72 vs. 60, P < 0.07). The colon tumor incidence was 56% in APCMin mice fed sucrose compared with 43% in those fed cornstarch, but this difference was not statistically significant. Mice consuming sucrose had significantly greater Ki-67 labeling index in colonic crypts compared to mice consuming cornstarch (37% vs. 32%, P <0.05), which indicated that mice consuming sucrose had a greater overall rate of cell proliferation in the colon. Mice consuming the high-sucrose diet had Significantly greater serum glucose concentrations compared to mice consuming cornstarch (10.09 :L- 0.57 vs. 8.08 :1: 0.57 mM, P < 0.05). Mice consuming sucrose tended to have higher serum insulin levels compared to mice consuming cornstarch (262.6 i 34.7 vs. 174.4 i 34.3 pmol/L, P = 0.07). Relative IGF-I mRNA expression in liver of mice fed sucrose was greater than that observed in mice fed cornstarch (0.166 vs. 0.092, p = 0.05). Hepatic mRNA expressions of IGF-II, IGF 8P1 and IGFBP3 were not influenced by diet. The objective of the second study was to further investigate alterations in global gene expression caused by APC gene mutation and by dietary carbohydrate source by using cDNA microarray analysis. Mice were assigned randomly at four weeks of age to one of two modified AIN-93G diets containing either 52.3% sucrose or cornstarch as the 154 sole carbohydrate source. Mice were fed these diets for 10 weeks, at which point they were sacrificed. Total RNA was extracted from epithelial cells scraped from the proximal third of the small intestine using Trizol reagent. cDNA synthesized using total RNA from small intestine was coupled with the fluorescent dyes Cy3 or Cy5 and hybridized on microarray slides printed with the NIA 15K gene set using a loop design. Intestinal expressions of 109 genes were significantly different between APCMin mice and wild-type mice, and many of them were associated with cell growth control and tumorigenesis (increased expressions of Clu, Ccnd2, Ccnbl, Btg4, Anxal, Gsptl; decreased expressions of Camkld, Septin 2, LatsZ, Dab2, Morf4ll). These observations indicate that APC gene mutations have a broad influence on expression of many genes associated with intestinal tumorigenesis. Expression of 306 genes was significantly influenced by dietary carbohydrate source. Among these genes, 87 were annotated and had expression altered by carbohydrate source by more than 50%. The expression patterns of genes altered by the high-sucrose diet suggest that sucrose altered the intestinal expression of genes in the insulin/IGF signaling pathway (increased expression of IGF-II and decreased expression of IGFBP3) in a manner consistent with elevated IGF Signaling in the intestinal epithelium. High-sucrose diets also changed expressions of genes associated with cell growth control and tumorigenesis (increased expressions of IGF2, PCNA, Csell, Idb2, Camk2g; decreased expressions of IGFBP3, Tial, Fancg, Bmprla, Cul4B), and the overall pattern of gene expression was consistent with the hypothesis that high dietary sucrose increases risk of intestinal tumorigenesis. 155 Dietary sucrose increased adenoma numbers in the proximal small intestine and cell proliferation in colon crypts in APCMin mice after lO-week of dietary treatment, but did not influence colonic adenoma development. We speculated that the lack of effect of sucrose on colonic adenoma development could be due to the relatively short time period (10 weeks) of dietary treatment for APCMin mice, which may not have been sufficient in duration for diets to exert their full impact on colon tumor progression. In the final study, the NSAID sulindac was added to all diets at 100 mg/kg to retard small intestinal tumorigenesis, thus allowing APCMin mice to live longer without significant morbidity from small intestinal adenomas. The objective of the third study was to utilize this extended-progression protocol to assess the impact of dietary carbohydrate and protein sources on colonic adenoma development and progression in APCMin mice. Mice were assigned randomly at four weeks of age to one of four modified AIN-93G diets containing different carbohydrate (cornstarch versus sucrose) and protein (soy versus casein) sources. Mice were fed these diets for 16 weeks. Mice consuming either sucrose or soy flour gained significantly more weight than those consuming cornstarch (P < 0.01) or casein (P < 0.05), respectively. Feeding sucrose promoted colon tumor development in APCMin mice compared with feeding cornstarch, with greater tumor incidence (59 vs. 30%, P < 0.01), greater tumor numbers (0.94 vs. 0.46 per mouse, P < 0.01) and greater tumor burden (6.8 vs. 2.5 mm3 per mouse, P < 0.01). Ki67 antigen immunohistochemistry indicated that mice fed sucrose had a higher epithelial cell proliferation rate in colon crypts (labeling index: 40 vs. 32%, P < 0.01; proliferative zone: 61% vs. 53%, P < 0.01), and reduced apoptosis (for TUNEL assay, apoptotic index: 3.8 156 vs. 5.6%, P < 0.05). Feeding diets containing soy flour promoted small intestinal adenoma development in APCMin mice compared with feeding casein, with increased sizes of adenomas (1.37 vs. 1.04 mmz, P < 0.01) and greater tumor burden (64 vs. 42 mm2 per mouse, P < 0.01). Dietary protein source had no effects on colon tumor development. Seven (7/35; 20% incidence) female mice consuming soy flour developed mammary gland tumors, whereas none (0/26; 0% incidence) of the female mice consuming casein-based diets developed mammary tumors (P < 0.05). In summary, diets containing high concentrations of sucrose significantly increased colon tumorigenesis in APCMin mice compared with cornstarch, and caused an increased rate of epithelial cell proliferation and decreased rate of programmed cell death in colonic crypts of APCMin mice. High-sucrose diets also altered gene expression patterns in intestinal epithelium that are associated with increased colon cancer risk. Increased risk for colon cancer caused by dietary sucrose in APCMin mice is associated with elevated insulin/IGF signaling in intestinal epithelium. Based on results of this research, fiIture research in three areas is of interest. First, the influence of dietary glucose and fructose on development of intestinal adenomas in APCMin mice needs to be further elucidated. In recent years, use of high fructose corn syrup (HFCS) as a sweetener in food and beverage has increased dramatically, so the potential influence of high intakes of HFCS on intestinal tumorigenesis is of particular interest. Second, further study on the role of hyperinsulinemia in promoting intestinal tumorigenesis should be pursued using animal models. For example, crossing APCMin mice with ob/ob mice, which carry a mutation in the leptin gene and become obese and exhibit diabetic-like symptoms early in life, would generate mice carrying both APC 157 gene and Lep°b gene mutations. If hyperinsulinemia directly stimulates intestinal tumorigenesis, mice carrying both defects would be expected to develop hyperinsulinemia and have increased risk for colon cancer compared with mice which carry APC gene mutations alone. This mouse model also could be used to test if dietary intervention, caloric restriction, or supplementation with recombinant leptin can correct hyperinsulinemia and therefore reduce risk for colon cancer. Finally, the results of gene expression profiling conducted as part of this research indicate that expression of many genes are modified by APC gene mutation or dietary carbohydrate source. Further research should be conducted to confirm these alterations in gene expression and explore the potential roles of these changes in intestinal tumorgenesis. 158 APPENDICES 159 Table A1. Genes up-regulated in small intestinal epithelium of mice consuming diets based on sucrose versus comstarchl. Clone ID Gene Name H3132808 hypothetical protein MGC38208 (MGC38208) H3131808 cadherin 2 (th2) H3111F06 Unknown H3051806 aldehyde reductase (aldose reductase)-like 6 (Aldrl6) H3041D10 expressed sequence AI327354 (AI327354) H3051C02 Unknown H3025803 stearoyl-Coenzyme A desaturase 2 (Scd2) H3 044H07 LOC241402 (LOC241402) H3088H02 Unknown H3147D07 Unknown H3 086D07 hypothetical Metallo-dependent hydrolases structure containing protein H3146802 IRON RESPONSIVE ELEMENT BINDING PROTEIN 1 IRE BP 1 IRON REGULATORY PROTEIN 1 IRPl F ERRITIN REPRESSOR PROTEIN ACONITATE HYDRATASE EC 4.2.1.3 CITRATE HYDRO LYASE H3 028F10 nuclear receptor coactivator 4 (Ncoa4) H3 023804 RIKEN cDNA 8230402H15 gene (8230402H15Rik) H3 068C01 Unknown H3 O70D01 expressed sequence C81457 (C81457) H3 1 l 8H06 inhibitor of DNA binding 2 (Idb2) H30 1 9H05 Unknown H3 1 5 5D05 similar to WD repeat domain 7 protein, isoform 1; TGF-beta resistance associated gene [Homo sapiens] (LOC208914) H3 095 H05 PSCD3 H3054E01 RAS p21 protein activator 3 (Rasa3) H3 1 49G02 MAD homolog 3 (Drosophila) (Madh3) H303 1 H12 SIMILAR T0 KIAA0971 PROTEIN (FRAGMENT). H3 1 321301 Unknown H3 076D03 RIKEN cDNA 3200002M13 gene (3200002M13Rik) 160 S/C2 Genebank ID 5.3 BG074237 5.2 BG074167 4.5 BG072515 4.1 BG067183 4.0 BG066309 3.9 BG067157 3.78G064900 3.6 BG066685 3.6 BG070577 3.6 BG075454 3.5 BG070351 3.5 BG075340 3.5 BG065226 3.4 BG064755 3.3 BG068683 3.3 BG068883 3 .3 80073126 3.3 8G064456 3.3 BG076077 3.2 8G071202 3.1 BG067440 3.1 BG075644 3.0 BG065510 2.9H3132801-5 2.9 BG069347 Table A1 (cont’d) H3159F10 Unknown H3046A08 Unknown H3032H12 Unknown H3071H07 Unknown H3041A02 RIKEN cDNA 2900053813 gene (290005 381 3Rik) H3021F12 proliferating cell nuclear antigen (Pena) H3082801 RIKEN cDNA 5730403810 gene (5730403810Rik) H3109F05 Unknown H3120008 Unknown H3100F08 membrane-associated protein 17 (Map17- pending) H3025805 serine/threonine kinase 3 (Ste20, yeast homolog) (Stk3) H3054804 LOC213321 (LOC213321) H3094D11 0DZ4 H3050F05 RETINOBLASTOMA PROTEIN INTERACTING ZINC FINGER PROTEIN RIz MTE BINDING PROTEIN MTB H3050E08 diaphorase 1 (NADH) (Dial) H3119H06 expressed sequence AI463719 (AI463719) H3135F02 SEC8 (S. cerevisiae) (Sec8) H3097H06 Unknown H3137812 Unknown H3045F02 SIMILAR T0 HY POTHETICAL PROTEIN F LJ20509 (F RAGMENT). H3099A03 Unknown H3050F09 Unknown H3053F08 Unknown H3114F10 CDNA FIS, CLONE , WEAKLY SIMILAR T0 RHO GTPASE ACTIVATING PROTEIN H3046H08 Unknown H3134F11 multiple endocrine neoplasia 1 (Menl) H3013D12 Unknown H3113H06 feminization l homolog b (C. elegans) (F emlb) H3096D08 Unknown 113001312 POLYCOMB-M33 INTERACTING PROTEIN RINGIB (FRAGMENT). 161 2.9 80076498 2.9 80066706 2.9 80065588 2.9 80069024 2.8 80066269 2.7 80064598 2.7CK334664 2.7 80072360 2.6 2.6 H3100F08-5 2.6 80064902 2.6 80067409 2.6 80071076 2.5 80067103 2.5 80067095 2.5 80073199 2.5 80074476 2.5 80071361 2.5 80074612 2.5 80066578 2.5 80071438 2.5 80067107 2.5 80067371 2.3 80072763 2.3 80066785 2.2 80074403 2.2 80063926 2.2 80072700 2.2 80071239 2.2 80063026 Table A1 (cont’d) H3076F12 REGULATOR OF 0 PROTEIN SIGNALING H3082D03 RIKEN cDNA 8430430L24 gene H3027A12 Unknown H3077806 long chain fatty acyl elongase (Lee-pending) H3077C06 RIKEN cDNA 4931430101 gene (493 1430101 Rik) H3095D03 Unknown H3142A01 RWl protein (Rwl-pending) H3145805 phosphate cytidylyltransferase 1, choline, alpha isoform (Pcytla) H3097D03 cyclin-dependent kinase inhibitor 1C (P57) (Cdknlc) H3132803 EST X83328 (X83328) H3107F05 RIKEN cDNA 2210415M14 gene (2210415M14Rik) H3033009 Unknown H3111H02 kallikrein 8, plasma 1 (Klkbl) H3057811 RIKEN cDNA 3110001D03 gene (31 10001 D03Rik) H3076D01 RIKEN cDNA 3110023802 gene (31 10023802Rik) H3 128812 hypothetical protein MGC19174 (MOC19174) H3142806 ZINC FINGER PROTEIN H3075C12 cysteine sulfinic acid decarboxylase (Csad) H3099F06 hypothetical Glutathione synthetase ATP- binding domain-like structure containing protein H3089F05 Unknown H3126H08 Unknown H3 13301 1 dihydrolipoamide S-acetyltransferase precursor (PDC-EZ) H3064804 Unknown H3044D12 Unknown H3002H07 RIKEN cDNA 1700019D03 gene (1700019D03Rik) H3038C01 PR8 MRNA CLEAVAGE COMPLEX II PROTEIN PCFll H3002D07 ribosomal protein S6 kinase polypeptide l (Rps6ka1) H3041D02 Unknown 162 2.2 80069379 2.2 80070065 2.2 80065075 2.1 80069509 2.1 80069521 2.1 80071159 2.1 80074995 2.] 80075254 2.180071313 2.1 80074232 2.1 80072177 2.0 80065662 2.0H3111H02-5 2.0 80067731 2.0 80069345 2.0H3128812-5 2.0 80075037 2.0 80069417 2.0 80071495 2.0 80070649 2.0H3126H08-5 2.0 80074331 2.0 80068347 1.9 80066644 1.9 80062997 1.9 80066040 1.9 80062954 1.9 H3041D02-5 Table A1 (cont’d) H3072A08 Similar to: similar to glyceraldehyde-3- phosphate dehydrogenase [Mus musculus] (LOC212429) H3001H10 THYMOSIN, BETA 10; PROTHYMOSIN BETA 10. H3100C10 Unknown H3001810 nucleolar and coiled-body phosphoprotein 1 (N olc 1) H3100D07 Unknown H3042F10 SIMILAR T0 RIKEN CDNA C08 H3031D03 aldolase 1, A isoform (Aldol) H3151D04 expressed sequence AW557061 (AW557061) H3033006 Unknown H3096D03 Unknown H303 1802 ALKYLDIHYDROXYACETONEPHOSPH ATE SYNTHASE, PEROXISOMAL PRECURSOR EC 2.5.1.26 ALKYL DI-IAP SYNTHASE ALKYLGLYCERONE PHOSPHATE H3098005 inhibitor of DNA binding 2 (Idb2) H3093D02 Similar to: similar to LAP-4 protein (Lymphoid nuclear protein related to AF 4) (LOC329124) H3097F04 RIKEN cDNA 5730469M10 gene (5730469M10Rik) H3019F06 tumor necrosis factor receptor superfamily, member 23 (Tnfrsf23) H3022C01 phosphate cytidylyltransferase 1, choline, alpha isoform (Pcytla) H3044A06 Unknown H3027D09 MARCKS-like protein (Mlp) H3032C02 SIMILAR T0 C01 49 H3079F02 Unknown H3046A01 RIKEN cDNA 2210008F15 gene (2210008F15Rik) H3085D03 phosphodiesterase 7A (Pde7a) H3048D04 RPBS-mediating protein (Rmp-pending) H3014H09 RIKEN cDNA 1110054A24 gene (1 110054A24Rik) H3085801 casein kinase 1, alpha 1 (Csnklal) 163 1.9 80069037 1.9 80063081 1.9H3100C10-5 1.9 80063054 1.9 80071554 1.9 80066409 1.9 80065457 1.9 80075762 1.9 80065659 1.8 80071234 1.8 80065433 1.8 80071421 1.8 80070978 1.8 80071335 1.8 80064436 1 .8 80064642 1.8 80066606 1.8 80065106 1.7 80065533 1.7CK334657 1.7 80066700 1.7 80070255 1.7 80066904 1.7 80064047 1.7 80070230 Table A1 (cont’d) H3044A02 SUCRASE ISOMALTASE, INTESTINAL [CONTAIN S: SUCRASE EC 3.2.1.48 ; ISOMALTASE 8C 3.2.1.- 10 H3124D09 phosphatidylinositol 3-kinase, C2 domain containing, alpha polypeptide (Pik3c2a) H3092012 CELL DIVISION CYCLE 2 RELATED PROTEIN KINASE 7 8C 2.7.l.- CDC2 RELATED PROTEIN KINASE 7 H3159804 Unknown H3029A05 hypothetical gene supported by NM_023565; AK011791 (LOC269412) H3076F01 natrium-phosphate cotransporter IIa C- terminal-associated protein 2 (AF 334612) H3084001 Unknown H3138F12 syndecan 4 (Sdc4) H3047H12 catenin alpha 1 (Catnal) H3062F11 Similar to: similar to hypothetical protein MOC955 [Homo sapiens] (8C035522) H3134D02 RIKEN cDNA 2610511017 gene (2610511017Rik) H3056C11 RIKEN cDNA 1110018L13 gene (1110018L13Rik) H3137C12 Similar to: 13 days embryo heart cDNA, RIKEN full-length enriched library, clonezD330024811 productzunknown EST, full insert sequence ' H3029012 Unknown H3101F11 tuftelin1(Tuftl) H3147806 Unknown H3006F12 growth factor receptor bound protein 7 (Grb7) H3048801 DNA segment, Chr 3, ERATO Doi 330, expressed (D38rtd330e) H3042D09 RIKEN cDNA 5730481H23 gene H3142C01 1500034806RIK H3023H03 Unknown H3060H05 Unknown H3076810 RIKEN cDNA 4432405K22 gene (4432405K22Rik) H3025D11 phosphofructokinase, liver, B-type H3036A12 Unknown H3147D09 RIKEN CDNA 2310061K06 gene (23 10061K06Rik) 164 1 .7 80066602 1.780073587 1.7 80070845 1.7H3159804-5 1.7 80065256 1.7 80069369 1.780070194 1.6 80074739 1.6 80066871 1.6 80068186 1.6 80074375 1.6 80067614 1.6 80074623 1.6 H3029012-5 1.6 80071659 1.680075431 1.6 80063390 1.6 80066882 1.6 80066389 1.6CK335109 1.6 1.6 80068020 1.6 80069332 1.6 80064930 1.6 80065861 1.6 H3147D09-5 Table A1 (cont’d) H3104803 ACTIN-ASSOCIATED PROTEIN PALLADIN (F RAGMENT). H3027C05 RIKEN cDNA 2400003C14 gene (2400003C14Rik) H3098F12 Similar to: Mus musculus, Similar to chromosome 17 open reading frame 31, clone IMAGEz5346419 H3100F02 Similar to: SplOO-rsl mRNA, partial sequence H3084809 Unknown H3126A04 insulin-like grth factor 2 (Igf2) H3009009 HYPOTHETICAL PROTEIN 8ST00098. H3019D05 Unknown H3074A11 Similar to CG8726 gene product (LOC218699) H3101C10 similar to glyceraldehyde-3-phosphate dehydrogenase [Mus musculus] (LOC237275) H3019806 hypothetical gene supported by 8C029161; 8C019633 (LOC268425) H3016Hll Similar to: cold shock domain protein A (Csda) H3039012 XRN l H3035D10 Unknown H3072A04 hypothetical Leucine-rich repeat, typical subtype containing protein H3062812 Unknown H3020D05 gene trap locus 1-13 H3083H04 Unknown H3030810 cathepsin B (Ctsb) H3085803 RNA binding motif protein 14 (Rbm14) H3004001 guanine nucleotide binding protein, alpha q polypeptide (Gnaq) H3056811 Unknown H3099009 calmodulin-like 4 (Calml4) H3041C02 2310030N02RIK H3029805 interleukin enhancer binding factor 2 (Ilf2) H3097H11 Unknown H3088A02 expressed sequence AA960436 (AA960436) H3015001 ubiquitin-conjugating enzyme 82A, RAD6 homolog (S. cerevisiae) (Ube2a) 165 1.680071905 1.5 80065091 1.580071418 1.5 80071570 1.5 80070144 1.5 80073613 1.5 80063647 1.5 80064417 1.5 80069222 1.5 80071626 1.5 80064425 1.5 80064216 1.5 80066178 1.4 80065801 1.4 80069033 1.4 80068141 1.4 80064494 1.4 800701 18 1.4 80065392 1.4 80070232 1.4 80063237 1.4 80067602 1.4 80071510 1.4 80066291 1.4 80065299 1.4H3097H11-5 1.4 80070496 1.4H3015001-5 Table A1 (cont’d) H3023804 hypothetical Thioredoxin-like structure 1.4 80064721 containing protein H3085001 IG MU CHAIN C REGION. 1.4 80070287 H3156A03 pericentriolar material 1 (Pcml) 1.4 80076129 H3044F11 Unknown 1.4 80066667 H3033D10 Unknown 1.4 80065629 113072812 LOC244840 (LOC244840) 1.3 80069052 H3054807 RIKEN cDNA 2810484M10 gene 1.3 80067412 (2810484M10Rik) H3132A02 melanoma antigen, family D, 2 (Maged2) 1.3 80074195 H3154811 RIKEN cDNA 9030425811 gene 1.3 80075988 (903042581 lRik) H3147H06 CALCYCLIN BINDING PROTEIN 1.3 80075491 (F RAGMENT). H3089804 ZINC FINGER PROTEIN 1.2 80070636 1mRNA expression of all the genes were significantly up-regulated by sucrose versus cornstarch (P < 0.05). 2S/C: intensity ratio, sucrose versus cornstarch. 166 Table A2. Genes down-regulated in small intestinal epithelium of mice consuming diets based on sucrose versus comstarch'. Clone ID Gene Name S/C2 Genebank ID H3028D11 40S RIBOSOMAL PROTEIN 0.6 80065203 H3002C12 5'-3' exoribonuclease 2 (Xrn2) 0.5 80062948 H3148805 60$ RIBOSOMAL PROTEIN 0.7H3148805-5 H3115012 60S RIBOSOMAL PROTEIN 0.6 80072862 H3004011 60S RIBOSOMAL PROTEIN L29. 0.7 80063246 H3042C03 adaptor-related protein complex AP-3, mu 1 0.7 80066375 subunit (Ap3m1) H3013804 adenylyl cyclase-associated CAP protein 0.3 80063930 homolog 1(Cap1) H3030809 AMINE OXIDASE [F LAVIN CONTAINING] 0.5 80065358 EC 1.4.3.4 MONOAMINE OXIDASE MAO H3029H01 ATP synthase mitochondrial F 1 complex 0.7 80065328 assembly factor 2 (Atpaf2) H3018810 basic leucine zipper and W2 domains 2 (82w2) 0.3 80064351 H3031808 bone morphogenetic protein receptor, type 1A 0.6 80065473 (Bmprla) H3145810 brain protein 16 (8rp16) 0.3 80075292 H3083806 cAMP responsive element binding protein 3 0.7 80070002 (Creb3) H3140A09 CARNITINE 0- 0.6 80074843 PALMITOYLTRANSFERASE I, MITOCHONDRIAL LIVER ISOFORM (EC 2.3.1.21) (CPT I) (CPTI-L) (FRAGMENT). H3106C05 CD2 antigen (cytoplasmic tail) binding protein 0.8 80072060 2 (Cd2bp2) H307SFO9 ciliary neurotrophic factor receptor (Cntfr) 0.4 80069461 H3043809 cullin 48 (Cul4b) 0.4 80066453 H3120F02 cytotoxic granule-associated RNA binding 0.5 80073264 protein 1 (Tial) H3015F06 DJ 126A5.1.2 (NOVEL DNAJ DOMAIN 0.680064114 PROTEIN) (ISOFORM 2) (F RAGMENT) homolog [Homo sapiens] H3148C09 EST AA792894 (AA792894) 0.6 80075525 H3025A05 eukaryotic translation initiation factor 3, 0.8 80064891 subunit 6 interacting protein (Eif336ip) H3118D09 expressed sequence AW548221 (AW548221) 0.7 80073078 H3106EO9 Fanconi anemia, complementation group G 0.4 80072083 (Fancg) 167 Table A2 (cont’d) H3102812 H3135011 H3116A10 H3009804 H31 14F01 H309981 1 H3125H07 H3125H11 H3075010 H3017EO3 H3050F12 H3122C11 H3085801 H3 069D02 H3085A01 H303 8809 H3075A09 H3021805 H3154007 H3087A12 H3043H09 H3122D09 H3096C05 H3016D12 H3031809 H3031A08 f-box and leucine-rich repeat protein 10 Fe receptor, IgG, low affinity III (F cgr3) FIBRONECTIN PRECURSOR (FN) (F RAGMENTS). flap structure specific endonuclease 1 (F enl) 01 to phase transition 1 (Gsptl) glutathione peroxidase 2, pseudogene 1 (pr2— psl) on chromosome 7 hemoglobin X, alpha-like embryonic chain in Hba complex (Hba-x) hemoglobin X, alpha-like embryonic chain in Hba complex (Hba-x) histone 4 protein (Hist4) HTPAP homolog [Homo sapiens] hydroxysteroid (l7-beta) dehydrogenase 4 (Hsd17b4) HY POTHETICAL 39.3 KDA PROTEIN (FRAGMENT). hypothetical EF-hand/Myb DNA binding domain/ELMZ domain containing protein hypothetical Glycerophosphoryl diester phosphodiesterase/Glycosyl hydrolase, starch- binding domain containing protein hypothetical Glycerophosphoryl diester phosphodiesterase/Glycosyl hydrolase, starch- binding domain containing protein hypothetical P-loop containing nucleotide triphosphate hydrolases structure containing protein hypothetical protein MGC 1 8894 (MGC 1 8894) hypothetical protein MOC37950 (MOC37950) insulin-like growth factor binding protein 3 (Igfbp3) lactotransferrin (Ltf) NADH CYTOCHROME 85 REDUCTASE nuclear distribution gene C homolog (Aspergillus) (Nude) nuclear receptor eo-repressor l (Ncorl) nucleolin (Nel) phospholipid scramblase 3 (Plscr3) polymerase (RNA) 11 (DNA directed) polypeptide 8 (25kDa) (Polr2e) 168 0.7 80071702 0.6 80074494 0.3 80072878 0.6 80063590 0.7 80072757 0.4 80071456 0.4 80073608 0.680073610 0.4 80069452 0.7 80064258 0.780067110 0.6 80073413 0.5 80070265 0.5 80068791 0.5 80070218 0.3 80066070 0.4 80069315 0.5 80064555 0.3 80076032 0.2 80070413 0.5 80066520 0.7 80073422 0.6 80071228 0.780064177 0.4 80065439 0.8 80065429 Table A2 (cont’d) H3114802 proteasome (prosome, macropain) 26S subunit, 0.7 80072746 non-ATPase, 7 (Psmd7) H3093809 PROTEIN DISULFIDE ISOMERASE A5 0.3 80070874 PRECURSOR EC 5.3.4.1 PROTEIN DISULFIDE ISOMERASE RELATED H3030808 PROTEIN KINASE C, TYPE EC 2.7.1.- 0.7 80065357 H3095006 RA827A, member RAS oncogene family 0.780071194 (Rab27a) H3035F09 Ras and a-factor-converting enzyme 1 0.3 80065824 homolog (Reel) H3093801 reticulon 4 (Rtn4) 0.6 80070989 H3139A08 Rho-guanine nucleotide exchange factor 0.5 80074770 (Rgnef) 113144809 ribosomal protein L6 (Rpl6) 0.5 80075206 H3010A10 ribosomal protein S11 (Rpsl 1) 0.780063667 H3006C11 ribosomal protein 818 (Rpsl8) 0.6 80063361 H3016811 ribosomal protein S18 (Rp818) 0.6 80064187 H3098009 ribosomal protein SS (RpsS) 0.6 H3098009-5 H3137809 RIKEN cDNA 0610007L05 gene 0.780074642 (0610007L05Rik) H3012C01 RIKEN cDNA 1110020104 gene 0.780063739 (1 1 10020104Rik) H3138A04 RIKEN cDNA 1300006C06 gene 0.8 80074679 (1300006C06Rik) H3015C05 RIKEN cDNA 2010320M17 gene 0.8 80064078 (2010320M17Rik) H3117D09 RIKEN cDNA 2210402A09 gene 0.7 80072996 (22 10402A09Rik) 83123005 RIKEN cDNA 2210407J23 gene 0.6 80073533 (2210407J23Rik) H3114805 RIKEN cDNA 2410127L17 gene 0.6 80072749 (2410127L17Rik) H3059C05 RIKEN cDNA 2610019N19 gene 0.4 80067882 (2610019N19Rik) H3121806 RIKEN cDNA 2900057D21 gene 0.5 80073343 (2900057D21Rik) H3057D02 RIKEN cDNA 6720463802 gene 0.7 80067712 (6720463802Rik) H3120D04 RNA BINDING 0.6 80073242 H3013C04 sequestosome 1 (qutml) 0.6 H3013C04-5 H3039A08 signal sequence receptor, delta (Ssr4) 0.5 80066111 H3001005 similar to CG9578 gene product (LOC226591) 0.7 80063069 169 Table A2 (eont’d) H3007A08 similar to Protein arginine N-methyltransferase 0.4 80063417 (LOC214572) H3055A09 SIMILAR T0 RIBOSOMAL PROTEIN S9 0.7 80067495 (UNKNOWN) (PROTEIN FOR MGC: 14341) (PROTEIN FOR MOC:2458) (PROTEIN FOR MOC:4138) homolog [Homo sapiens] H3076009 SIMILAR T0 SPLICING FACTOR, 0.6 80069388 ARGININE/SERINE-RICH 7 (35KD). H3083C07 Similar to: defensin related cryptdin 3 (Defcr3) 0.3 80069980 H3093001 Similar to: Mus musculus, clone 0.6 80071009 IMAGE:4456744, partial eds H3053007 Similar to: serine/threonine kinase 13 0.180067380 (aurora/IPL-like) (Stk13) H3008A02 solute carrier family 25 (mitochondrial carrier; 0.6 80063501 oxoglutarate carrier), member 11 (Slc25a11) H3084A08 SPECKLE TYPE POZ 0.6 80070133 H3122C07 STATHMIN (PHOSPHOPROTEIN P19) 0.4 BG073409 (PP19) (ONCOPROTEIN 18) (OP18) (LEUKEMIA-ASSOCIATED PHOSPHOPROTEIN P18) (PP17) (PROSOLIN) (METABLASTIN) (PR22 PROTEIN) (LEUKEMIA-ASSOCIATED GENE PROTEIN). H3014C08 surfeit gene 4 (Surf4) 0.3 80063996 H3142D08 TIP120 FAMILY PROTEIN 0.6 80075030 H3086804 transcriptional regulator, SIN3A (yeast) 0.7 80070358 (Sin3a) H3082H12 ubiquitin conjugating enzyme 6 (ch6p- 0.7 80069952 pending) H3140806 ubiquitin-conjugating enzyme 82L 3 (Ube213) 0.5 H3140806-5 H3119005 WD REPEAT PROTEIN 0.680073188 H3014D06 ZINC FINGER PROTEIN 219 homolog [Mus 0.780064004 musculus] H3101C05 4930488P06RIK 0.7 80071621 H3095C04 AU067695 0.880071149 H3068A02 D10JHU828 0.4 80068660 H3049805 EIF285 0.7 80067005 H3114F05 HBA-Al 0.5 80072760 H3062801 L3M8TL3 0.4 80068130 H3089A01 MAPILC3 0.7 80070588 H3037F09 NCORl 0.5 80065991 H3037D12 Unknown 0.2 80065971 170 Table A2 (cont’d) H3037C10 H3107802 H3044008 H3050003 H3040002 H3055C01 H3133805 H3091010 H3137A08 H3034808 H3089805 H3051012 H3095003 H3097A01 H3100H05 H3040A1 1 H3101809 H3133A01 H3062802 H3100A12 H3039A02 H3001A06 H303 8A03 H3065A12 H3092F02 H3094001 H3095A07 H31 15802 H3120A09 H3135C08 H3137804 H3139808 H3 145C 1 0 Unknown Unknown Unknown Unknown Unknown Unknown Unknown Unknown Unknown Unknown Unknown Unknown Unknown Unknown Unknown Unknown Unknown Unknown Unknown Unknown Unknown Unknown Unknown Unknown Unknown Unknown Unknown Unknown Unknown Unknown Unknown Unknown Unknown 0.2 80065957 0.3 H3107802-5 0.3 80066675 0.3 800671 12 0.3 80066250 0.3 80067510 0.3 80074309 0.4 80070926 0.4 80074597 0.4 80065727 0.4 80070604 0.4 80067212 0.4 80071 191 0.4 H3097A01-5 0.4 80071590 0.4 H3040Al 1-5 0.4 80071648 0.5 H3133A01-5 0.5 80068131 0.5 H3100A12-5 0.5 80066106 0.6 0.6H3038A03-5 0.6 80068401 0.7 H3092F02-5 0.780071 102 0.7 80071 131 0.6 80072832 0.6 80073213 0.6 80074448 0.6 80074604 0.6 H3139808-5 0.6 1mRNA expression of all the genes were significantly down-regulated by sucrose versus cornstarch (P < 0.05). 2S/C: intensity ratio, sucrose versus cornstarch. 171 Table A3. Genes up-regulated in small intestinal epithelium of APCMin mice versus wild-type mice'. Clone ID Gene Name M/W2 Genebank ID H3051H08 8-cell translocation gene 4 (Btg4) 14.9 80067220 H3034806 Unknown 10.1 H3034806-5 H3143005 Unknown 7.9 80075141 H3108A04 clusterin (Clu) 5.6 80072209 H3051C05 Similar to: Mus musculus, Similar to hypothetical protein 4.9 80067159 MOC5576, clone MOC154819 IMAGE:6308771, complete eds H3146F09 similar to chromosome 22 open reading frame 3 [Homo 4.4 80075392 sapiens] (LOC279765) H3002801 selenoprotein R (Sepr) 3.7 80062960 H3071H01 Unknown 3.7 80069019 H3159003 TISSUE FACTOR PATHWAY INIHBITOR PRECURSOR 3.6 H3159003-5 (TFPI) (LIPOPROTEIN- ASSOCIATED COAGULATION INHIBITOR) (LACI) (EXTRINSIC PATHWAY INHIBITOR) (8P1). H3064H11 Unknown 3.6 80068390 H3154D09 hypothetical protein MOC11659 (MGCl 1659) 3.5 80076005 H3096806 Unknown 3.5 80071222 H3108A05 hypothetical protein MOC28622 (MGC28622) 3.4 80072210 H3074003 Unknown 3.3 80069286 H3106808 Similar to: similar to Synaptotagmin XI (SthI) 3.2 80072082 (LOC229521) H3146H06 weakly similar to CTL2 PROTEIN [Homo sapiens] 3.1 80075411 H3030D02 Unknown 3.1 80065374 H3054H08 RIKEN cDNA 1110033009 gene (1110033009Rik) 3.1 80067483 H3109808 expressed sequence AA987064 (AA987064) 3.1 80072315 H3040D06 Unknown 3.0 80066222 H3112D06 RIKEN cDNA 2610027Ll6 gene (2610027L16Rik) 3.0 H3112D06-5 H3034F08 SIMILAR T0 RHO GUANINE NUCLEOTIDE 2.9 80065738 EXCHANGE FACTOR (GEF) 3. H3148F06 HY POTHETICAL ZINC FINGER PROTEIN 2.9 80075555 H3105810 cyclin 81, related sequence 1 (Ccnbl-rsl) 2.9 80071965 H3071A01 Unknown 2.9 H3142H06 general transcription factor 11 H, polypeptide 1 (62kD 2.7 80075067 subunit) (0tf2h1) H3130F08 Similar to: Mus musculus, clone IMA08:3497440 2.7 80074083 H3092810 ADP-ribosylation-like 3 (Arl3) 2.6 80070958 172 Table A3 (eont’d) H3096F12 Unknown 2.6 80071263 H3098810 similar to KIAA0475 gene product (LOC215015) 2.6 80071392 H3144812 Unknown 2.5 80075179 H3087A12 lactotransferrin (Ltf) 2.5 80070413 H3146806 Unknown 2.5 80075344 H3124808 Unknown 2.4 80073563 H3147C02 GRIP ASSOCIATED PROTEIN l 2.4 80075438 H3159807 Unknown 2.4 80076410 H3051012 Unknown 2.4 80067212 H3150D10 cleavage and polyadenylation specificity factor 1 (Cpsfl) 2.4 80075692 H3143H10 Unknown 2.4 80075157 H3154F06 upstream transcription factor 1 (Usfl) 2.4 H3154F06-5 H3036A12 Unknown 2.4 80065861 H3148F04 Unknown 2.4 80075553 H3008A10 MYB binding protein (P160) la (Mybbpla) 2.3 80063508 H3102H08 expressed sequence AU041483 (AU041483) 2.3 80071761 H3149H02 Unknown 2.3 80075654 H3128812 hypothetical protein MOC19174 (MOC19174) 2.3 H3128812-5 H3060810 Unknown 2.3 H3060810-5 H3107D05 RIKEN cDNA 1110004C05 gene (1110004C05Rik) 2.3 80072156 H3152D10 expressed sequence AIl82287 (AIl82287) 2.2 80075841 H3029801 solute carrier family 11 (proton-coupled divalent metal ion 2.2 80065264 transporters), member 2 (Slc11a2) H3095F12 Unknown 2.2 80071188 H3144012 glypican 1 (Gpcl) 2.2 80075228 H3138A07 T-complex expressed gene 1 (Tcel) 2.1 80074682 #Num! ARA9 2.1 H3127811 ubiquitously expressed transcript (Uxt) 2.0 80073796 H3121F05 hypothetical Lipocalins structure containing protein 2.0 80073353 H3064803 mitogen-activated protein kinase 6 (Mapk6) 2.0 CK334608 H3142810 Unknown 2.0 80075050 H3158D03 protein tyrosine phosphatase, non-receptor type 18 (Ptpn18) 2.0 80076324 H3008H01 annexin A1 (Anxal) 2.0 80063567 H3088F03 protein tyrosine phosphatase 4a3 (Ptp4a3) 1.9 80070554 H3096F 01 Unknown 1.9 H3096F 01-5 H3041D06 FALSE P73 TARGET 1.9 80066305 H3020F08 Similar to: Wiskott-Aldrich syndrome homolog (human) 1.9 80064517 (Was) 173 Table A3 (cont’d) H3099811 glutathione peroxidase 2, pseudogene 1 (pr2-psl) on 1.9 80071456 chromosome 7 H3098009 ribosomal protein SS (RpsS) 1.9 H3098009-5 H3144806 H19 FETAL LIVER MRNA. 1.9 80075175 H3152F10 Nedd4 WW binding protein 4 (N4wbp4-pending) 1.9 80075859 H3012H04 similar to R29144_1 [Homo sapiens] 1.8 80063794 H3106C05 CD2 antigen (cytoplasmic tail) binding protein 2 (Cd2bp2) 1.8 80072060 H3120D04 RNA BINDING 1.8 80073242 H3030H10 high mobility group box 3 (ngb3) 1.8 80065420 H3141808 Unknown 1.8 80074966 H3097H03 extracellular proteinase inhibitor (Expi) 1.8 80071358 H3137A10 hypothetical Dbl domain (dbl/cdc24 rhOGEF family) 1.8 80074599 containing protein H3049F12 60S RIBOSOMAL PROTEIN 1.8 80067024 H3118H11 SMALL NUCLEAR RIBONUCLEOPROTEIN 1.8 80073131 POLYPEPTIDE G. H3002H04 Similar to: UDP-GlcNAczbetaGal beta-1,3-N- 1.8 80062994 acetylglucosaminyltransferase l (83gnt1), mRNA H3012H06 expressed sequence AA408877 (AA408877) 1.8 80063884 H3118803 PLASMA MEMBRANE CALCIUM TRANSPORTING 1.8 80073084 ATPASE EC 3.6.3.8 PLASMA MEMBRANE CALCIUM PUMP ISOFORM PLASMA MEMBRANE CALCIUM ATPASE ISOFORM H3132801 ZINC FINGER PROTEIN 1.8 80074240 H3012D10 WHSCI . 1.8 BG063759 H3152D01 cyclin D2 (Ccnd2) 1.8 80075832 H3008BO3 RIKEN cDNA1200003M11 gene (1200003M11Rik) 1.7 BG063512 H3158H01 SERINE/THREONINE PROTEIN PHOSPHATASE 5 (EC 1.7 BG076363 3.1.3.16) (PPS) (PROTEIN PHOSPHATASE T) (PPT). H3141D09 Unknown 1.7 80074957 H3097F12 Unknown 1.7 80071343 H3158H12 RIKEN cDNA 2310047M15 gene (2310047M15Rik) 1.7 80076372 H3050D06 SH3-binding domain glutamic acid-rich protein (Sh3bgr) 1.7 80067083 H3098F12 Similar to: Mus musculus, Similar to chromosome 17 open 1.7 80071418 reading frame 31, clone IMAGE:5346419 H3088D12 weakly similar to MOLYBDOPTERIN SYNTHASE 1.6 80070540 SULFURYLASE [Homo sapiens] H3137F04 Unknown 1.6 80074647 174 Table A3 (cont’d) H31 12D08 H3065A12 H3148801 H3031808 H3125803 H3025812 H3014F10 H3013A04 H31 14F01 H3120D03 H3 1 02F 01 H3101A11 H3010A10 H3102F10 H3024D04 H3081H11 H31 18004 H3026D04 H3125Hl 1 H3145C10 H3152002 H3156802 H3009C1 1 H3145D1 1 H3059F01 H3096802 H3122D09 H3013D1 l 26S PROTEASOME NON ATPASE REGULATORY SUBUNIT 11 268 PROTEASOME REGULATORY SUBUNIT S9 26S PROTEASOME REGULATORY SUBUNIT P44 Unknown inhibitor of grth family, member 1 (Ingl) bone morphogenetic protein receptor, type 1A (Bmprla) Unknown hypothetical protein MGC19174 (MGC19174) CEROID LIPOFUSCINOSIS NEURONAL PROTEIN 5 CLNS f-box only protein 6b (Fbxo6b) 01 to phase transition 1 (Gsptl) SIMILAR T0 TEMO CLATHRIN COAT ASSEMBLY PROTEIN AP17 (CLATHRIN COAT ASSOCIATED PROTEIN AP17) (PLASMA MEMBRANE ADAPTOR AP-2 17 KDA PROTEIN) (HA2 17 KDA SUBUNIT) (CLATHRIN ASSEMBLY PROTEIN 2 SMALL CHAIN). spindlin (Spin) ribosomal protein S1 1 (Rpsl 1) ZINC FINGER PROTEIN BINDING . proteasome (prosome, macropain) 26S subunit, non- ATPase, 8 (Psmd8) ribosomal protein S17 cyclin D2 (Ccnd2) hemoglobin X, alpha-like embryonic chain in Hba complex (Hba-x) Unknown hypoxanthine guanine phosphoribosyl transferase (Hprt) YIPPEE PROTEIN (FRAGMENT) homolog [Homo sapiens] 1.6 1.6 1.6 1.6 1.6 1.6 1.6 1.6 1.6 1.6 1.6 1.6 1.5 1.5 1.5 1.5 1.5 1.5 1.5 1.5 1.5 1.5 EUKARYOTIC TRANSLATION INITIATION FACTOR 21.5 SUBUNIT 2 EUKARYOTIC TRANSLATION INITIATION FACTOR 2 BETA SUBUNIT 81F 2 ribosomal protein S24 (Rps24) ubiquitin carboxy-terrninal hydrolase L1 (Uchll) Unknown nuclear distribution gene C homolog (Aspergillus) (Nude) METALLOTHIONEIN-II (MT-II). 175 1.5 1.5 1.5 1.5 1.5 80072569 80068401 80075539 80065473 80073745 80064909 80064027 80063892 80072757 80073241 80071735 CK334730 80063667 80071744 80064834 80070030 80073104 80065010 80073610 80075862 80076174 80063 608 80075284 80067910 H3096802-5 80073422 80063925 Table A3 (cont’d) H3151008 PEPTIDYLPROLYL ISOMERASE MATRIN CYP (BC 1.5 80075790 5.2.1.8) (PEPTIDYLPROLYL CIS- TRANS ISOMERASE) (MATRIN CYCLOPHILIN) (MATRIN CYP) (PPIASE) homolog [Rattus norvegicus] H3092F02 Unknown 1.4 H3092F02-5 H3108803 histocompatibility 13 (H13) 1.4 80072220 H3111A06 ribosomal protein L22 (Rp122) 1.4 80072480 H3013C04 sequestosome 1 (qutrnl) 1.4 H3013C04-5 H3041F11 Unknown 1.4 80066329 H3029H01 ATP synthase mitochondrial F1 complex assembly factor 2 1.4 80065328 (Atpaf2) H309581] Unknown 1.3 H3095811-5 H3134D02 RIKEN cDNA 2610511017 gene (2610511017Rik) 1.3 80074375 H3155H04 hypothetical ARM repeat structure containing protein 1.3 80076118 H3135C08 Unknown 1.3 80074448 H3001D02 heparan sulfate (glucosamine) 3-0-sulfotransferase l 1.3 80063038 (Hs3stl) H3084010 Similar to: 13 days embryo male testis cDNA, RIKEN full- 1.3 80070203 length enriched library, clonez6030477023 produetzunclassifiable, full insert sequence H3138A04 RIKEN cDNA 1300006C06 gene (1300006C06Rik) 1.3 80074679 H3022F07 TROPOMYOSIN CHAIN 1.2 80064681 ImRNA expressions of all the genes were significantly up-regulated in APC?Vlln mice vs. wild-type mice (P < 0.05). 2M/W: intensity ratio, Min versus wild-type. 176 Table A4. Genes down-regulated in small intestinal epithelium of APCMin mice versus wild-type mice‘. Clone ID Gene Name M/W2 Genebank ID H3055804 Unknown H3150805 GATA binding protein 2 (Gata2) H3074A09 PROBABLE N 2 ,N 2 DIMETHYLGUANOSINE TRNA METHYLTRANSFERASE EC 2.1.1.32 TRNA GUANINE 26,N 2 N 2 METHYLTRANSFERASE TRNA 2,2 DIMETHYLGUANOSINE 26 METHYLTRANSFERASE TRNA M 2,2 026 H3108804 8D4_MOUSE Ubiquitin-protein ligase Nedd-4 H3046C05 Unknown H3088C08 Unknown H3155D07 homeodomain interacting protein kinase 3 (Hipk3) H3147C06 Unknown 83042801 Unknown H3104010 8030025C11R1K H3053H04 Unknown H3063Ell Unknown H3044804 DNA segment, Chr 13, ERATO Doi 275, expressed (D13Ertd275e) H3038801 JAK3_MOUSE Tyrosine-protein kinase JAK3 (Janus kinase 3) (JAK-3) H3048A03 Unknown H3099D07 hypothetical Serine/threonine specific protein phosphatase containing protein H3068807 expressed sequence AI467391 (AI467391) H3034003 Unknown H3073A08 Unknown H3080D07 UBINUCLEIN homolog [Homo sapiens] H3003C07 MUC2_RAT Mucin 2 precursor (Intestinal mucin 2) H3053C05 HYPOTHETICAL 46.8 KDA PROTEIN. H3055C07 Unknown H3063F07 CALRETICULIN PRECURSOR CRP55 CALREGULIN HACBP H3047C06 Unknown 177 0.1 80067537 0.1H3150805-5 0.1 80069220 0.2 80072221 0.2 80066727 0.2 80070525 0.2 80076079 0.2 80075441 0.2 80066391 0.2 80071931 0.2 80067387 0.2 80068267 0.2 80066648 0.2 80066063 0.2 H3048A03-5 0.2 80071474 0.2 80068712 0.2 80065745 0.2 80069128 0.2 80069812 0.3 80063119 0.3 80067336 0.3 80067516 0.3 80068274 0.3 80066817 Table Amont’d) H3073803 Similar to: proton-dependent high affinity oligopeptide transporter (Pept2) gene, complete eds H3074011 RIKEN cDNA 5730403J10 gene (5730403J 10Rik) H3105F08 nucleoporin 62 (Nup62) H3076H09 hypothetical protein MGC40770 (MGC40770) H3121007 weakly similar to hypothetical protein DKFZp434D193.1 (fragment) [Homo sapiens] H3026809 RIKEN cDNA 2810460C24 gene (28 10460C24Rik) H3053805 AV028368 H3111H02 kallikrein 8, plasma 1 (Klkbl) H3109A04 receptor (calcitonin) activity modifying protein 2 H3055F05 D88RTD4578 H31 12006 transforming growth factor beta 1 induced transcript 4 (Tgfbli4) H3112C04 Unknown H3030A09 feminization 1 homolog a (C. elegans) (Femla) H3026H05 D11MOH45 H3043F09 gene rich cluster, C8 gene (Grce8) H3032804 kinesin family member 58 (Kibe) H3056A09 inositol 1,4,5-triphosphate receptor 2 (Itpr2) H3092C10 Similar to: Mus musculus, clone IMAGE:4010812 H3076807 ZINC FINGER PROTEIN H3088D05 dystonin (Dst) H305 1H12 RETINOBLASTOMA PROTEIN INTERACTING ZINC FINGER PROTEIN RIZ MTE BINDING PROTEIN MT 8 H3032A09 peroxisomal integral membrane protein (Pmp47) H3158802 RIKEN cDNA 5730555F13 gene H3084D03 UDP-GLCNAC:A- l ,3-D-MANNOSIDE 8- 1,4-N- ACETYLGLUCOSAMINYLTRANSFERASE IV (EC 2.4.1.145) homolog [Homo sapiens] H3105001 ring finger protein 178 0.3 80069134 0.3 80069294 0.3 80072008 0.3 80069400 0.3 80073367 0.3 80065025 0.3 80067358 0.3 H3111H02-5 0.3 80072299 0.3 80067550 0.3 80072602 0.3 80072603 0.3 80065347 0.3 80072605 0.3 80072606 0.3 80072607 0.3 80072608 0.3 80072609 0.3 80072610 0.4 8007261 1 0.4 80072612 0.4 80072613 0.4 80072614 0.4 80072615 0.4 80072616 Table A4 (eont’d) H305 7803 hypothetical Prokaryotic membrane lipoprotein lipid attachment site containing protein H303 8809 hypothetical P-loop containing nucleotide triphosphate hydrolases structure containing protein H3046D03 ITGAV H3115811 protein kinase C, lambda (Prkcl) H3098F06 SERINE/THREONINE PROTEIN PHOSPHATASE 2A, 56 KDA REGULATORY SUBUNIT, EPSILON ISOFORM (PP2A, B SUBUNIT, 8' EPSILON ISOFORM) (PP2A, 8 SUBUNIT, 856 EPSILON ISOFORM) (PP2A, 8 SUBUNIT, PR61 EPSILON ISOFORM) (PP2A, 8 SUBUNIT, R5 EPS H3035805 Unknown H307001 1 Unknown H3076F 12 REGULATOR 0F 0 PROTEIN SIGNALING H3041F12 Unknown H3 141006 hypothetical Acyl-COA N-acyltransferases (Nat) structure containing protein H3121C02 follistatin-like (Fstl) H3046808 hypothetical Homeobox domain containing protein H3062809 PROTEIN TYROSINE PHOSPHATASE 4A1. H3050D09 similar to claspin [Homo sapiens] (LOC277677) H3072C03 Unknown H3041A02 RIKEN cDNA 2900053813 gene (2900053813Rik) H3157006 Unknown H3097F03 Similar to: Mus musculus, clone IMAGE:5011849 H3123A06 Unknown H3035801 TR8-3 (TR8-3) H3057808 Unknown H3116C04 SEC8 (S. cerevisiae) (Sec8) H31 1 1H06 Unknown H3096811 Unknown H3022C06 Similar to: hypothetical protein L00208146 (LOC208146) 179 0.4 80072617 0.4 80072618 0.4 80072619 0.4 80072620 0.4 80072621 0.4 80072622 0.4 80072623 0.4 80072624 0.4 80072625 0.4 80072626 0.4 80072627 0.4 80072628 0.4 80072629 0.4 80072630 0.4 80072631 0.4 80072632 0.4 80072633 0.4 80072634 0.4 80072635 0.4 80072636 0.4 80072637 0.4 80072638 0.4 80072639 0.4 80072640 0.4 80072641 Table A4 (eont’d) H3032811 RAD26L HYPOTHETICAL PROTEIN, ALTERNATIVELY SPLICED PRODUCT, SIMILAR T0 (AF217319) PUTATIVE REPAIR AND RECOMBINATION HELICASE RAD26L (FRAGMENT) homolog [Homo sapiens] H3085C02 Traf and Tnf receptor associated protein H3080003 ribnuclease inhibitor H3132A02 melanoma antigen, family D, 2 (Maged2) H3131F06 developmentally regulated GTP binding protein 1 (Drgl) H3065C07 Unknown H3035F03 Unknown H3035809 hypothetical protein MGC46970 (MGC46970) H3025F02 8C030932 H3001D04 HISTONE H2A.G (H2A/G) (H2A.3). H3098GO2 Unknown H3085805 RIKEN cDNA 5730533P17 gene (5730533P17Rik) H3058805 RIKEN cDNA 1110008L16 gene H3041F09 solute carrier family 2, (facilitated glucose transporter), member 8 (Slc2a8) H3127F08 RIKEN cDNA 5530600P05 gene H3068F07 Unknown H303 5004 peptidase 4 (Pep4) H3044805 Unknown H3040810 81NG4 protein (8ing4) H3025006 2810410P22RIK H3046C09 2310035N15RIK H3095H11 Unknown H3103810 MACGAP homolog [Homo sapiens] H3008004 GATA binding protein 2 (Gata2) H31 12012 Unknown H3096F03 Unknown H3026C09 ubiquitin specific protease 23 H3098A11 Unknown H3024001 mini chromosome maintenance deficient 5 (S. cerevisiae) (MemdS) H3122006 neuropilin (Nrp) H3050C07 Unknown 180 0.4 80072642 0.4 80072643 0.4 80072644 0.4 80072645 0.4 80072646 0.4 80072647 0.4 80072648 0.4 80072649 0.4 80072650 0.4 8007265 1 0.5 80072652 0.5 80072653 0.5 80072654 0.5 80072655 0.5 80072656 0.5 80072657 0.5 80072658 0.5 80072659 0.5 80072660 0.5 80072661 0.5 80072662 0.5 80072663 0.5 80072664 0.5 80072665 0.5 80072666 0.5 80072667 0.5 80072668 0.5 80072669 0.5 80072670 0.5 80072671 0.5 80072672 Table A4 (cont’d) H3097F04 RIKEN cDNA 5730469M10 gene (5730469M10Rik) H3138806 lethal giant larvae homolog (nglh) H3132F06 pleiomorphie adenoma gene-like 2 (Plag12) H3120H09 Unknown H3018D01 thyroid hormone receptor interactor 4 (Trip4) H3070810 SIMILAR T0 RIKEN CDNA N24 H3070H05 Unknown H3026808 human ubiquitin H3102Cll Unknown H3121002 RIKEN cDNA 3100004P22 gene (3100004P22Rik) H3120008 Unknown H3140006 Mpv17 transgene, kidney disease mutant (Mpv17) H3030A08 replication protein Al (70 kDa) (Rpal) H3001A06 Unknown H3036004 Unknown H3116F04 SIMILAR T0 REV/REX ACTIVATION DOMAIN BINDING PROTEIN H3112807 3 KETOACYL COA THIOLASE, MITOCHONDRIAL EC 2.3.1.16 BETA KETOTHIOLASE ACETYL COA ACYLTRANSFERASE MITOCHONDRIAL 3 0XOACYL COA H3112808 Unknown H3120H12 Unknown H3076F01 natrium-phosphate cotransporter IIa C- tenninal-associated protein 2 (AF 334612) H3024D09 WD repeat domain 1 (Wdrl) H306581 1 Unknown H3098A08 RIKEN cDNA 1010001M04 gene (1010001M04Rik) H3026807 sideroflexin 1 (Sfxnl) H3012010 bisphosphate 3'-nueleotidase 1 (Bpntl) H3056C11 RIKEN cDNA 1110018L13 gene (1110018L13Rik) H3101C09 Unknown H3048C04 testis expressed gene 189 H3068C01 Unknown H3027A12 Unknown 181 0.5 80072673 0.5 80072674 0.5 80072675 0.5 0.5 80064330 0.5 80068903 0.5 80068934 0.5 H3026808-5 0.5H3102C11-5 0.5 80073362 0.5 0.5 80074905 0.5 80065346 0.5 0.5 80065911 0.5 80072928 0.5 80072552 0.5 80072580 0.5 80073295 0.5 80069369 0.5 80064839 0.5 80068443 0.5 80071372 0.5 80065024 0.5 80063789 0.5 80067614 0.5 80071625 0.5 80066894 0.5 80068683 0.5 80065075 Table A4 (eont’d) H3047A01 hypothetical RabGAP/TBC domain containing protein H3097A12 Unknown H3120801 CADHERIN H3056A11 Ngfi-A binding protein 1 (Nabl) H3035003 Unknown H3026FO3 RIKEN cDNA 5730536A07 gene (5730536A07Rik) H3112010 disabled homolog 2 (Drosophila) (Dab2) H3067A12 RIKEN cDNA 2410003L22 gene (2410003L22Rik) H3116C08 Unknown H3023006 microsomal triglyceride transfer protein (Mttp) H3050809 RIKEN cDNA 2900054P12 gene (2900054P12Rik) H3 044A01 Unknown H3008A03 interleukin 17 receptor (Ill7r) H3112008 exportin 4 (Xpo4-pending) H3140801 Unknown H3068807 RIKEN cDNA 4833425H18 gene (4833425H18Rik) H3068C04 DEAD (aspartate-glutamate-alanine-aspartate) box polypeptide 3 (Ddx3) H3082806 LIM-DOMAIN PROTEIN LMP-l homolog [Rattus norvegicus] H3070A10 nuclear receptor co-repressor 1 (N eorl) H3026A12 Unknown H3089810 Unknown H3049A01 SUMO 1 SPECIFIC PROTEASE 1 EC 3.4.22.- SENTRIN SPECIFIC PROTEASE SENP6 PROTEASE H3142C01 1500034806RIK H3075A10 RIKEN cDNA 2810489L22 gene (2810489L22Rik) H3088C03 expressed sequence AU015605 (AU015605) 83016003 hypothetical protein KIAA0610 (fragment) homolog [Homo sapiens] H3124D09 phosphatidylinositol 3-kinase, C2 domain containing, alpha polypeptide (Pik3c2a) H3099801 Similar to: 7 days neonate cerebellum cDNA, RIKEN full-length enriched library, clone:A730047D01 product:unknown EST 0.5 80066790 0.5 H3097A12-5 0.6 80073251 0.6 80067591 0.6 80065830 0.6 80065031 0.6 80072606 0.6 80068579 0.6 80072898 0.6 80064780 0.6 80067096 0.6 80066601 0.6 80063502 0.6 80072604 0.6 80074877 0.6 80068677 0.6 80068686 0.6 80070079 0.6 80068856 0.6H3026A12-5 0.6 80070642 0.6 80066959 0.6CK335109 0.680069316 0.6 80070520 0.6 80064200 0.6 80073587 0.6 80071447 182 Table A4 (eont’d) H3088A02 expressed sequence AA960436 (AA960436) H3065810 SPIR 2 H3027D02 MYO INOSITOL MONOPHOSPHATASE H3007A09 SET translocation H3007F08 poliovirus receptor-related 3 (Pvrl3) H3046A01 RIKEN cDNA 2210008F15 gene (2210008F15Rik) H3020012 Unknown H3076A10 Unknown H3109010 KUNITZ-TYPE PROTEASE INHIBITOR 2 PRECURSOR (HEPATOCYTE GROWTH FACTOR ACTIVATOR INHIBITOR TYPE 2) (HAl-2). H3101C11 RIKEN cDNA 4921528807 gene (4921528807Rik) H3062F1 1 Similar to: similar to hypothetical protein MGC955 [Homo sapiens] (8C035522) H3097H1 1 Unknown H3152811 septin 2 (Sept2) H3053D11 Unknown H3095A05 ESRI H3039C09 pMS protein (PmS-pending) H3077F04 OSBPL8 H3022D11 protein phosphatase 4, regulatory subunit 1 (PPP4r1) H3046A09 F K506 binding protein 10 (65 kDa) (F kbplO) H3133A08 Unknown H3133A12 zinc finger protein 94 (pr94) H3052808 similar to DKFZP56400823 protein (L00231440) H3035F09 Ras and a-factor-converting enzyme 1 homolog (S. cerevisiae) (Reel) H3111C09 aminolevulinic acid synthase 1 (Alasl) H3087801 annexin A4 (Anxa4) H3023D04 hypothetical Integral membrane protein, DUF6 containing protein H3146006 aconitase 2, mitochondrial (Ae02) H3034804 RCL SIMILAR T0 PUTATIVE C MYC H3 069A 1 0 Unknown H3033809 similar to Herrnansky-Pudlak syndrome protein variant (LOC276962) 183 0.6 80070496 0.6 80068442 0.6 80065100 0.6 80063418 0.6 80063474 0.6 80066700 0.6 80064531 0.6 80069486 0.6 80072373 0.6 80071627 0.6 80068186 0.6H3097H11-5 0.6 80075848 0.6 80067352 0.6 80071 129 0.6 80066132 0.6 80069555 0.6 80064662 0.6 80066707 0.6 80074270 0.6 80074273 0.6 80067276 0.680065824 0.6H3111C09-5 0.6 80070448 0.6 80064744 0.6 80075401 0.6 80065723 0.6 80068763 0.6 80065639 Table A4 (eont’d) H3149C10 triosephosphate isomerase (Tpi) H3056801 actin related protein 2/3 complex, subunit 5 (165 kDa) (ArpcS) H3110C01 erythrocyte protein band 4.1-like 4b (pr4.ll4b) H3015C01 B-cell receptor-associated protein 31 (8eap3l) H3021A12 tryptophanyl-tRNA synthetase (Wars) H3091C03 Unknown H3092002 Similar to: Mus musculus, clone IMAGE:4010812 H3011805 Unknown H3100005 Unknown H3139801 heat shock 70kD protein 8 H3088805 galactosidase, alpha (Gla) H3054807 RIKEN cDNA 2810484M10 gene (2810484M10Rik) H3077801 Similar to: X-linked lymphocyte-regulated 5 (Xlr5) H3079C11 ECGFl H3063002 espin (Espn) H3144A09 AUTOSOMAL HIGHLY CONSERVED H3129806 expressed sequence AW546258 (AW546258) H3015001 ubiquitin-conjugating enzyme 82A, RAD6 homolog (S. cerevisiae) (Ube2a) H3068A08 kinesin-like 5 (mitotic kinesin-like protein 1) (KnslS) H3098804 Unknown H3014005 RIKEN cDNA 0610038P07 gene (0610038P07Rik) H3062812 Unknown H3001H02 Unknown H3098003 STROMAL INTERACTION MOLECULE H3095C11 Unknown H3060H05 Unknown H3042C03 adaptor-related protein complex AP-3, mu 1 subunit (Ap3m1) H3016811 ANKYRIN H3032C02 SIMILAR T0 C01 49 H301 8001 DEAD (aspartate-glutamate-alanine-aspartate) box polypeptide 3 (Ddx3) H3099009 calmodulin-like 4 (Calml4) 184 0.6 80075608 0.7 80067628 0.7 8007241 1 0.7 80064074 0.7 80064551 0.7 80070794 0.7CK334685 0.7 80063851 0.780071582 0.7 80074803 0.7 80070545 0.780067412 0.7 80069540 0.7 80069714 0.7 80068280 0.7 80075168 0.7 80073952 0.7 H3015001-5 0.7 80068666 0.7 80071402 0.7 80064033 0.780068141 0.7 0.7H3098GO3-5 0.7H3095C11-5 0.7 80068020 0.7 80066375 0.7 80064159 0.780065533 0.7 80064364 0.780071510 Table A4 (eont’d) H3042C09 weakly similar to GASC-l PROTEIN [Homo sapiens] H3116812 AL024097 H3052C12 serine/threonine kinase 11 (Stkl 1) H3062C11 SET domain, bifurcated l (Setdbl) H3079A09 cyclin A2 (Ccna2) H3041C02 2310030N02RIK 83078003 IMPORTIN BETA 2 SUBUNIT KARYOPHERIN BETA 2 SUBUNIT TRANSPORTIN M9 REGION INTERACTION PROTEIN H3060C01 INTERLEUKIN l RECEPTOR ASSOCIATED KINASE EC 271- IRAK H3085005 SIMILAR T0 H3024011 hypothetical Ubiquitin earboxyl-terrninal hydrolase family 2 containing protein H3141D02 EFNAS H3042A07 stromal cell derived factor 4 (Sdf4) H3095809 Similar to: similar to mastigoneme-like protein [Chlamydomonas reinhardtii] (LOC277139) H3138811 sema domain, immunoglobulin domain (Ig), transmembrane domain (TM) and short cytoplasmic domain, (semaphorin) 4G (Sema4g) H3089005 collagen, type IV, alpha 3 (Goodpasture antigen) binding protein (Col4a3bp) H3027C05 RIKEN cDNA 2400003C14 gene (2400003C14Rik) H3069C09 Rho-associated coiled-coil forming kinase 1 (Rockl) H3003803 Unknown H3147H06 CALCYCLIN BINDING PROTEIN (FRAGMENT). ImRNA expressions of all the genes were significantly down-regulated in APCMin mice vs. wild-type mice (P < 0.05). 2M/W: intensity ratio, Min versus wild-type. 185 0.7 80066379 0.7 80072924 0.7 80067258 0.7 80068152 0.7 80069688 0.7 80066291 0.7 H3 078003-5 0.7 80067964 0.7 80070291 0.7 80064874 078007495] 0.7 80066355 0.7 80071 174 0.7 80074727 0.7 80070661 0.880065091 0.8 80068786 0.8 80063137 0.8 80075491 Table A5. Genes whose expressions were altered differently by APC genotype and dietary sucrose”. Clone ID Gene Name M_C W_C M_S W_S Genebank 1D H3113003 AGRIN 4.1 2.2 2.3 2.980072686 H3031801 ATP BINDING CASSETTE, 0.8 0.6 0.5 0.880065432 SUB FAMILY F, MEMBER 2 IRON INHIBITED ABC TRANSPORTER 2 HUSSY H3145810 brain protein16(8rp16) 0.1 0.0 0.0 0.080075292 H3147H06 CALCYCLIN BINDING 10.0 18.2 18.2 17.980075491 PROTEIN (FRAGMENT). H3106C05 CD2 antigen (cytoplasmic tail) 0.2 0.2 0.2 0.180072060 binding protein 2 (Cd2bp2) H3025A05 eukaryotic translation initiation 1.5 1.0 0.9 1.180064891 factor 3, subunit 6 interacting protein (Eif356ip) H3088A02 expressed sequence AA960436 0.1 0.3 0.2 0.380070496 (AA960436) H3003810 GDPMANNOSE 0.4 0.8 0.8 0.680063142 PYROPHOSPHORYLASE H3072A04 hypothetical Leucine-rieh 0.3 0.7 0.7 0.680069033 repeat, typical subtype containing protein H3038809 hypothetical P-loop containing 0.2 0.5 0.0 0.280066070 nucleotide triphosphate hydrolases structure containing protein H3104812 hypothetical protein 0.6 0.3 0.4 0.580071880 MGC31247 (MGC31247) H3037A07 hypothetical TPR repeat 0.5 0.1 0.1 0.280065934 containing protein H3024011 hypothetical Ubiquitin 1.0 1.8 1.2 1.180064874 carboxyl-terrninal hydrolase family 2 containing protein H3132A02 melanoma antigen, family D,2 0.2 0.7 0.4 0.780074195 (Maged2) H3084802 MYOSIN PHOSPHATASE 6.1 11.9 12.1 5.680070139 H3089004 neuregulin4(Nrg4) 3.2 4.4 4.3 3.980070660 186 Table A5 (cont’d) H3075810 H3 008003 H3154809 H3121C06 H3076F12 H3138A04 H3085806 H3031802 H3054807 H3050809 H3152811 H3158H01 H3062C1 1 H3074Al 1 H3015810 NON KINASE CDC42 8F F ECTOR PROTEIN proteasome (prosome, macropain) subunit, beta type 7 (Psmb7) PROTEIN KINASE CLK3 (EC 2.7.1.-) (CDC-LIKE KINASE 3). recombination activating gene 1 gene activation (Rga) REGULATOR OF G PROTEIN SIGNALING RIKEN cDNA 1300006C06 gene (1300006C06Rik) RIKEN cDNA 1810003N24 gene (1810003N24Rik) RIKEN cDNA 2810410M20 gene (2810410M20Rik) RIKEN cDNA 2810484M10 gene (2810484M10Rik) RIKEN cDNA 2900054P12 gene (2900054P12Rik) septin 2 (Sept2) SERINE/THREONINE PROTEIN PHOSPHATASE 5 (EC 3.1.3.16) (PPS) (PROTEIN PHOSPHATASE T) (PPT). SET domain, bifurcated 1 (Setdbl) similar to CG8726 gene product (LOC21 8699) similar to HIPPOCALCIN- LIKE I [Mus musculus] 187 3.4 2.9 2.3 1.3 0.0 4.0 0.5 2.1 4.1 1.2 0.7 1.5 1.0 2.5 4.4 5.9 2.5 5.3 0.8 0.3 2.6 0.6 1.1 10.0 2.8 1.7 0.5 2.6 3.7 2.8 5.0 2.0 5.9 0.9 0.3 2.6 0.7 1.2 9.1 1.6 1.1 0.6 2.2 4.7 2.9 4.2 80069327 2.9 80063558 4.5 80075986 1.3 80073320 0.5 80069379 2.5 80074679 0.5 80070281 1.6 80065467 9.6 80067412 2.0 80067096 1.3 80075848 0.7 80076363 1.9 80068152 4.4 80069222 3.480064107 Table ASLcont’d) H3099801 H3101F 1 1 H3012807 H3025H08 H3041C02 H3042805 H3060H05 H3062812 H3064D1 l H3089D05 H3093A08 H3096802 H3101C05 H310201 1 H31 12012 H3145C10 H3156H09 Similar to: 7 days neonate cerebellum cDNA, RIKEN full-length enriched library, clonezA730047D01 product:unknown EST, fiIll insert sequence tuftelin 1 (Tuftl) Unknown Unknown Unknown Unknown Unknown unknown unknown unknown unknown unknown unknown unknown unknown unknown unknown 1.2 0.2 5.2 4.5 0.4 0.0 0.2 0.4 8.8 0.2 1.4 3.0 1.6 0.5 0.6 5.1 0.3 3.3 2.4 0.6 0.7 7.8 9.1 7.3 8.0 0.9 0.9 0.1 0.2 0.6 0.6 1.2 1.2 12.7 12.8 0.2 0.1 3.0 3.0 1.5 1.7 1.3 0.9 1.7 1.1 2.4 1.3 2.6 2.6 0.1 0.1 2.7 80071447 0.6 80071659 6.3 H3012807-5 6.7 80064969 0.9 80066291 0.1 H3042805-5 0.6 80068020 1.1 80068141 10.0 80068342 0.4 80070626 2.4 80070862 1.7 H3096802-5 1.1 80071621 1.3 H3102C11-5 1.7H3112012-5 2.4 0.2 8007621 1 1Data represent normalized mean intensity values of mRNA expression for the corresponding gene in the treatment groups: M_C (APCMm mouse consuming cornstarch), W_C (wild-type mouse consuming cornstarch), M_S (APCMm mouse consuming sucrose), W_S (wild-type mouse consuming sucrose). 2Significant interaction (P < 0.05) between effects of diet and genotype. 188 REFERENCE 189 Adenis A, Peyrat JP, Heequet 8, Delobelle A, Depadt G, Quandalle P, Bonneterre J, Demaille A (1995) Type I insulin-like growth factor receptors in human colorectal cancer. Eur J Cancer 31A:50—55. Altsehuler DL, Ribeiro—Neto F (1998) Mitogenic and oncogenic properties of the small 0 protein Raplb. Cell Biology 95(13):7475-9. American Cancer Society (2004) Cancer Facts & Figures. American Institute of Cancer Research/World Cancer Research Fund (AICR/WCRF) (1997) Food, Nutrition and the Prevention of Cancer: A Global Perspective. Baker SJ, Reddy EP (1998) Modulation of life and death by the TNF receptor superfamily. Oncogene 1 7:3261-70. 8ames S, Grubbs C, Setchell KD & Carlson J (1990) Soybeans inhibit mammary tumors in models of breast cancer. Prog. Clin. Biol. Res. 347:239-253. Beavo JA (1995) Cyclic nucleotide phosphodiesterases: functional implications of multiple isoforms. Physio Rev 75:725. Bedi A, Pasricha PJ, Akhtar AJ, Barber, JP, Bedi, GC, Giardiello, FM, Zehnbauer, BA, Hamilton, SR and Jones, RJ (1995) Inhibition of apoptosis during development of colorectal cancer. Cancer Res 55: 1 81 1-1816. Behrens J, von Kries JP, Kilhl M Bruhn L, Wedllich D, Grosschedl R, Birchmeier W (1996) Functional interaction of B-catenin with transcription factor Lef-l. Nature 382:638-42. Behrens P, Brinkmann U, Wellmann A (2003) CSEIL/CAS: its role in proliferation and apoptosis. Apoptosis 8(1):39-44. Benito 8, 0brador A, Stiggelbout A (1990) A population-based case-control study of colorectal cancer in Majorca: dietary factors. Int J Cancer 45:69-76. Benito E, Cabeza 8, Moreno V, 0brador A (1993) Diet and colorectal adenomas: a case- control study in Majorca Int J Cancer 55:213-166. Bennink MR, 0m AS, Miyagi Y (1999) Inhibition of colon cancer by soy flour but not by genistein or a mixture of isoflavones. . F ASEB J 13:A50. Bennink MR., 0m AS. and Miyagi Y (2000) Inhibition of colon cancer by wheat bran, soy flour and flax. FASEB J 14:A217. 190 Bertling E, Hotulainen P, Mattila PK, Matilainen T, Salminen M, Lappalainen P (2004) Cyclase-assoeiated protein 1 (CAPl) promotes cofilin-induced actin dynamics in mammalian nonmuscle cells. Mol Biol Cell 15(5):2324-34. Bertram MJ, Berube NG, Hang-Swanson X, Ran Q, Leung JK, Bryce S, Spurgers K, Bick RJ, Baldini A, Ning Y, Clark L], Parkinson 8K, Barrett JC, Smith JR, Pereira-Smith 0M (1999) Identification of a gene that reverses the immortal phenotype of a subset of cells and is a member of a novel family of transcription factor-like genes. Mol Cell Biol 19(2): 1479-85. Bessesen DH (2001) The role of carbohydrates in insulin resistance. . J Nutr 131227828- 868. Bidoli E, Franceschi S, Talamini R (1992) Food consumption and cancer of colon and rectum in north-eastem Italy. Int J Cancer 50:223-9. Boland CR, Montgomery CK, Kim YS (1982) Alterations in human colonic mucin occurring with cellular differentiation and malignant transformation. . Proc Natl Acad Sci USA 79(6):2051-5. Bostick RM, Potter JD, Kushi LH, Sellers TA, Steinmetz KA, McKenzie DR, Gapstur SM, Folsom AR (1994) Sugar, meat, and fat intake, and non-dietary risk factors for colon cancer incidence in Iowa women (United States). Cancer Causes Control 5(1):38-52. Bravo R. (1986) Synthesis of the nuclear protein cyclin (PCNA) and its relationship with DNA replication. Exp Cell Res 163:287-93. Bristol J8, Emmett PM, Heaton KW, Williamson RCN (1985) Sugar, fat, and risk of colon cancer. Br Med J 291 :1467-71. Bray GA, Nielsen SJ, Popkin BM (2004) Consumption of high-fructose corn syrup in beverages may play a role in the epidemic of obesity. Am J Clin Nutr 79(4):537-43. Burkitt DP (1969) Related disease-related cause? Lancet 2: 1229-3 1. Busserolles I, Rock 8, Gueux 8, Mazur A, Grolier P, Rayssiguier Y (2002) Short-term consumption of a high-sucrose diet has a pro-oxidant effect in rats. Br J Nutr 87(4):337- 42. Bristol JB, Emmett PM, Heaton KW, Williamson, RCN (1985) Sugar, fat, and risk of colon cancer Br Med J 291:1467-71.Caderni G, Dolara P, Spagnesi T, Lucemi C, Bianchini F, Mastrandrea V, Morozzi G (1993) Rats fed high starch diets have lower colonic proliferation and faecal bile acids than high sucrose-fed controls. J Nutr 123:704- 12. 191 Cademi G, Bianchini F, Mancina A, Spagnesi MT, Dolara P (1991) Effect of dietary carbohydrates on the growth of dysplastic crypt foci in the colon of rats treated with 1,2- dirnethylhydrazine. Cancer Res 51(14):3721-5. Cademi G, Luceri C, Spagnesi MT, Giannini A, Biggeri A, Dolara P (1994) Dietary carbohydrates modify azoxymethane-induced intestinal carcinogenesis in rats. . J Nutr 124(4):517-23. Cademi G, Luceri C, Lancioni L, Dolara P (1996) Dietary sucrose, glucose, fructose, and starches affect colonic functions in rats. Nutr Cancer 25(2):179-86. Cademi G, Lancioni L, Luceri C, Giannini A, Lodovici M, Biggeri A, Dolara P (1997) Dietary sucrose and starch affect dysplastic characteristics in carcinogen-induced aberrant crypt foci in rat colon. . Cancer Lett 114:39-41. Cassidy A, Bingham SA, Cummings JH (1994) Starch intake and colorectal cancer risk: an international comparison. . Br J Cancer 69(5):937-42. Centonze S, Boeing H, Leoci C, Guerra V, Miseiagna G (1994) Dietary habits and colorectal cancer in a low risk area Results fi'om a population-based case-control study in Southern Italy. Nutr Cancer 21 :233-46. Chatterjee S. (1999) Neutral sphingomyelinase: past, present and future. Chem Phys Lipids 102(1-2):79-96. Chen T, Yang 1, Irby R, Shain KH, Wang HG, Quackenbush J, Coppola D, Cheng JQ, Yeatman TJ (2003) Regulation of caspase expression and apoptosis by adenomatous polyposis coli. . Cancer Res 63(15):4368-74. Chiu CH, McEntee MF, Whelan J (1997) Sulindac causes rapid regression of preexisting tumors in Min mice independent of prostaglandin biosynthesis. . Cancer Res 57:4267-73. Clemmons DR, Klibanski A, Underwood L8, McArthur J W, Ridgway EC, Beitins IZ, Van Wyk JJ (1981) Reduction of immunoreactive somatomedin-C during fasting in humans. J Clin Endocrinol Metab 53:1247-50. Corpet DE, Peiffer G, Tache S (1998) Glycemie index, nutrient density, and promotion of aberrant crypt foci in rat colon. Nutr Cancer 32(1):29-36. Crane R, Gadea 8, Littlepage L, Wu H, Ruderrnan JV (2004) Aurora A, meiosis and mitosis. Biol Cell 96(3):215-29. Crawford HC, F ingleton BM, Rudolph-Owen LA, Goss KJ, Rubinfeld 8, Polakis P (1999) The metalloproteinase matrilysin is a target of beta-catenin transactivation in intestinal tumors. . Oncogene 18:2883-91. 192 Cubbage ML, Suwanichkul A, Powell DR (1990) Insulin-like growth factor binding protein-3. Organization of the human chromosomal gene and demonstration of promoter activity. J Biol Chem 265(21):12642-9. Cumming JH, Bingham SA, Heaton KW, Eastwood MA (1992) Fecal weight, colon cancer and dietary intake of NSP (dietary fibre). . Gastroenterology 103: 1783-89. Curmi PA, Gavet 0, Charbaut E, Ozon S, Lachkar-Colmerauer S, Maneeau V, Siavoshian S, Maueuer A, Sobel A (1999) Stathmin and its phosphoprotein family: General properties, biochemical and functional interaction with tubulin. Cell Struct Funct 24(5):345-57. Davies CG, Netto FM, Glassenap N, Gallaher CM, Labuza TP, Gallaher DD (1998) Indication of the Maillard reaction during storage of protein isolates J Agrie Food Chem 46: 2485-89. Davies MJ, Bowey EA, Adlcrcrwutz H, Rowland IR, Rumsby PC (1999) Effects of soy or rye supplementation of high fat diets on colon tumor development in azoxymethane- treated rats. . Carcinogenesis 20:927-31. Dely M (2003) Sugars, insulin sensitivity, and the postprandial state. Am J Clin Nutr 78(4):865S-8728. de Rooij J, Boenink NM, van Triest M, Cool RH, Wittinghofer A, 808 JL (1999) PDZ- GEF l, a guanine nucleotide exchange factor specific for Rapl and Rap2. J Biol Chem 274(53):38125-30. D'Errico A, Garbisa S, Liotta LA, Castronovo V, Stetler-Stevenson WG, Grigioni WF (1991) Augmentation of type IV collagenase, laminin receptor, and K167 proliferation antigen associated with human colon, gastric, and breast carcinoma progression. Mod Pathol 4(2):239-46. Deschner EE (1977) Experimental induced cancer of the colon. . Cancer 34:824-8. Deschner 88, Long FC (1977) Colonic neoplasms in mice produced with six injections of 1,2-dimethyhydrazine. . Oncology 34:255-7. Deschner 88, Raieht RF (1981) Kinetic and morphologic alterations in the colon of a patient with multiple polyposis. . Cancer 47(10):2440-5. De Stefani 8, Mendilaharsu M, Dcneo-Pellegrini H (1998) Sucrose as a risk factor for cancer of the colon and rectum: a case-control study in Uruguay. . Int J Cancer 75(1):40- 4. 193 Dixon DA, Balch GC, Kedersha N, Anderson P, Zimmerman GA, Beauchamp RD, Prescott SM (2003) Regulation of cyclooxygenase-2 expression by the translational silencer TIA-1. J Exp Med 198(3):475-81. Dohlman HG, Thomer J (1997) RGS Proteins and Signaling by Heterotrimeric 0 Proteins. J Biol Chem 272:3871-4. Dragsted L0, Daneshvar B, Vogel U, Autrup HN, Wallin H, Risom L, Moller P, Molck AM, Hansen M, Poulsen HE, Loft S (2002) A sucrose-rich diet induces mutations in the rat colon. Cancer Res 62(15):4339-45. Dunn SE, Kari F W, French J, Leininger JR, Travlos 0, Wilson R, Barrett JC (1997) Dietary restriction reduces insulin-like growth factor I levels, which modulates apoptosis, cell proliferation, and tumor progression in p53-deficient mice. . Cancer Res 57:4667-72. Dunnigan MG, Fyfe T, McKiddie MT, Crosbie SM (1970) The effects of isocaloric exchange of dietary starch and sucrose on glucose tolerance, plasma insulin and serum lipids in man. . Clin Sci 38:1—9. Elliott SS, Keim NL, Stern J S, Teff K, Havel PJ (2002) Fructose, weight gain, and the insulin resistance syndrome. . Am J Clin Nutr 76(5):911-22. Eisenhauer PB, Harwig SS, Lehrer RI. (1992) Cryptdins: antimicrobial defensins of the murine small intestine. Infect Immun 60(9):3556-65. Englyst HN, Kingman SM, Cummings JH (1992) Classification and measurement of nutritionally important starch fractions. . Eur J Clin Nutr 46(2):S33-50. Faber E (1995) Cell proliferation as a major risk factor for cancer: A concept of doubtful validity. . Cancer Res 55: 3759-62. Fearon 8R, Vogelstein BA (1990) Genetic model for colorectal tumorigenesis. . Cell 61: 759-67. FEMS (Federation of European Microbiological Societies) (1987) Microbiol Ecol 95:163. Fioravanti L, Cappclletti V, Miodini P, Ronchi 8., Brivio M & Di Fronzo G (1998) Genistein in the control of breast cancer cell growth: insights into the mechanism of action in vitro. Cancer Lett 130:143-152. F oulds L (1958) The natural history of caneer.J Chronic Dis 8(1):2-37. F oulstone 8, Prince S, Zaceheo 0, Burns JL, Harper J, Jacobs C, Church D, Hassan AB (2005) Insulin-like growth factor ligands, receptors, and binding proteins in cancer. J Pathol 205(2): 145-53. 194 Franceschi S, Dal Maso L, Augustin L, Negri E, Parpinel M, Boyle P, Jenkins DJ, La Vecchia C (2001) Dietary glycemic load and colorectal cancer risk. . Ann Oncol 2(2):173-8. Fritz WA., Coward L, Wang J & Lamartiniere CA (1998) Dietary genistein: perinatal mammary cancer prevention, bioavailability and toxicity testing in the rat. Carcinogenesis 19:2151-58. F ukuma M, Okita H, Hata J, Umezawa A (2003) Upregulation of Id2, an oncogenie helix-loop-helix protein, is mediated by the chimeric EWS/ets protein in Ewing sarcoma. Oncogene 22: 1-9. Furuuchi K, Tada M, Yamada H. (2000) Somatic mutations of the APC gene in primary breast cancers. American J Pathol 156:1997-2005. Gerdes J (1990) Ki-67 and other proliferation markers useful for immunohistological diagnostic and prognostic evaluations in human malignancies. Semin Cancer Biol 1(3): 199-206. Gerdes J, Li L, Schlueter C, Duchrow M, Wohlenberg C, Gerlach C, Stahmer I, Kloth S, Brandt 8, F lad HD (1991) Irnmunobioehemical and molecular biologic characterization of the cell proliferation-associated nuclear antigen that is defined by monoclonal antibody Ki-67. Am J Pathol 138(4):867-73. Giardiello FM, Hamilton. SR, Kush AJ, Piantadosi S, Hylind LM, Celano P, Brooker SV, Robinson CR, Offerhaus GJ (1993) Treatment of colonic and rectal adenomas with sulindac in familial adenomatous polyposis. . N Engl J Med 328:1313-16. Giovannucci E (1995) Insulin and colon cancer. . Cancer Causes Control 6: 164-79. Giovannucci 8, Pollak MN, Platz EA, Willett WC, Stampfer MJ, Majeed N, Colditz GA, Speizer F8, Hankinson SE (2000) A prospective study of plasma insulin-like grth factor-1 and binding protein-3 and risk of colorectal neoplasia in women. . Cancer Epidemiol Biomarkers Prev 9(4):345-9. . Giovannucci 8 (2001) Insulin, insulin-like growth factors and colon cancer: a review of the evidence. . J Nutr 131(11):3109S-20S. Goldberg Y, Nassif II, Pittas A, Tasai LL, Dynlacht 8D, Rigas 8, Shiff SJ (1996) The anti-proliferative effect of sulindac and sulidac sulfide on HT-29 colon cancer cells: Alterations in tumor suppressor and cell-regulatory proteins. . Oncology 12:893-901. Hague A, Paraskeva C (1995) The short-chain fatty acid butyrate induces apoptosis in colorectal tumour cell lines. . Eur J Cancer Prev 4(5):359-64. 195 Hakam A, Yeatman TJ, Lu L, Mora L, Marcet G, Nicosia SV (1999) Expression of insulin-like growth factor-1 receptor in human colorectal cancer. Hum Pathol 30:1128— 33. Hakkak, R., Korourian, S., Shelnutt, S. R., Lensing, S., Ronis, M. J. & Badger, T. M. (2000) Diets containing whey proteins or soy protein isolate protect against 7,12- dimethylbenz(a)anthracene-induced mammary tumors in female rats. Cancer Epidemiol Biomark Prev 9:113-117. Hakkak R, Korourian S, Ronis MJJ, Johnston JM, Badger TM (2001) Soy protein isolate consumption protects against azoxymethane-induced colon tumors in male rats. . Cancer Lett 166: 27-32. - Hall PA, Woods AL (1990) Immunohistochemical markers of cellular proliferation: achievements, problems and prospects. Cell Tissue Kinet 23(6):505-22. Hardman, W.E., Cameron, I.L., Heitrnan, D.W. and Contreras, 8. Demonstration of the need for endpoint validation of putative biomarkers: Failure of aberrant crypt foci to predict colon cancer incidence. Cancer Res., 51, 6388-6392, 1991. Hallberg LM, Ikeno Y, Englander E, Greeley 0H Jr (2000) Effects of aging and caloric restriction on IGF-I, IGF-I receptor, IGFBP3 and IGFBP-4 gene expression in the rat stomach and colon. . Regul Pept 89(1-3):37-44. Hanif R, Pittas A, Feng Y, Koutsos MI, Qiao L, Staiano-Coico L, Shiff SI, Rigas 8 (1996) Effects of nonsteroidal anti-inflammatory drugs on proliferation and on induction of apoptosis in colon cancer cells by a prostaglandin-independent pathway. . Biochem Pharmacol 52:237-45. Hassan AB, Howell JA (2000) Insulin-like gowth factor II supply modifies growth of intestinal adenoma in Ape(Min/+) mice. . Cancer Res 60:1070-6. Hauge H, Patzke S, Delabie J, Aasheim HC (2004) Characterization of a novel immunoglobulin-like domain containing receptor. Biochem Biophys Res Commun 323(3):970-8. Havel PJ (2002) Control of energy homeostasis and insulin action by adipocyte hormones: leptin, acylation stimulating protein, and adiponectin. . Curr Opin Lipidol 13:51—9. Hawrylewicz EJ, Huang HH. & Blair WH. (1991) Dietary soybean isolate and methionine supplementation affect mammary tumor progression in rats. J Nutr 121:1693- 98. Hawrylewicz EJ, Zapata JJ & Blair WH. (1995) Soy and experimental cancer: animal studies. J Nutr 125:698S-708S. 196 He TC, Sparks AB, Rago C, Hermeling H, Zawel L, da Costa LT (1998) Identification of c-MYC as a target of the APC pathway. . Science 281:1509-12. He XC, Zhang J, Tong W0, Tawfik 0, Ross J, Seoville DH, Tian Q, Zeng X, He X, Wiedemann LM, Mishina Y, Li L (2004) BMP signaling inhibits intestinal stem cell self- renewal through suppression of Wnt-beta-catenin signaling. Nat Genet 36(10):]117-21. Hegde P, Qi R, Abernathy R, Gay C, Dharap S, Gaspard R, Earle-Hughes I, Snesrud E, Lee NH, and QuackenbushJ. (2000) Biotechniques 29(3):548-62. Hsieh CY, Santell RC, Haslam SZ & Helferich W0 (1998) Estrogenic effects of genistein on the growth of estrogen receptor-positive human breast cancer (MCF-7) cells in vitro and in vivo. Cancer Res 58:3833-3 838. Hisamatsu T, Watanabe M, Ogata H, Ezaki T, Hozawa S, Ishii H, Kanai T, Hibi T. (1999) Interferon-inducible gene family 1-8U expression in colitis-associated colon cancer and severely inflamed mucosa in ulcerative colitis. Cancer Res 59(23):5927-31. Ho K, Kalle W, Le THS, Lam W, Tang C (1999) Reduced expression ofAPC and DCC gene protein in breast cancer. Histopathology 35:249—56. Hofbauer LC, Rafferzeder M, Janssen 08, Gartner R (1995) Insulin-like growth factor I messenger ribonucleic acid expression in porcine thyroid follicles is regulated by thyrotropin and iodine. Eur J Endocrinol 132:605—610. Holmes MD, Pollak MN, Willett WC, Hankinson SE (2002) Dietary correlates of plasma insulin-like growth factor I and insulin-like growth factor binding protein 3 concentrations Cancer Epidemiol Biomarkers Prev 11(9):852-61. Holt PR, Moss SF, Kapetanakis AM, Petrotos A, Wang S (1997) Is Ki-67 a better proliferative marker in the colon than proliferating cell nuclear antigen? Cancer Epidemiol Biomarkers Prev 6(2):131-5. Hong J, Zhang G, Dong F, Rechler MM (2002) Insulin-like growth factor (IGF)-binding protein-3 mutants that do not bind IGF-I or IGF-II stimulate apoptosis in human prostate cancer cells. J Biol Chem 277(12):]0489-97. Howe, GR (1992) Dietary intake of fiber and decreased risk of cancer 8 of the colon and rectum: Evidence from combined analysis of thirteen case control studies. J Natl Cancer Inst 84: 1887-1895. Howe JR, Bair JL, Sayed MG, Anderson M8, Mitros FA, Petersen GM, Velculescu V8, Traverso G, Vogelstein B (2001) Gennline mutations of the gene encoding bone morphogenetic protein receptor 1A in juvenile polyposis. Nat Genet 28(2): 1 84-7. 197 Hu F8, Manson JE, Liu S, Hunter D, Colditz GA, Michels KB, Speizer F 8, Giovannucci E (1999) Prospective study of adult onset diabetes mellitus (type 2) and risk of colorectal cancer in women. .J Natl Cancer Inst 91: 542-7. Hu JF, Vu TH, Hoffman AR (1996) Promoter-specific modulation of insulin-like growth factor II genomic imprinting by inhibitors of DNA methylation.J Biol Chem 271(30):18253-62. IARC (International Agency for Research on Cancer, Lyon, France) (2002) http://www- depdbiarcfr/globocan/GL080framehtm. Ilyas M, Straub J, Tomlinson 1PM, Bodmer WF (1999) Genetic pathways in colorectal and other cancers. . European Journal of Cancer 35:1986-2002. Jacoby RF, Marshall DJ, Newton MA, Novakovic K, Tutsch K, Cole C8, Lubet RA, Kelloff GJ, Verma A, Moser AR, Dove WF (1996) Chemoprevention of Spontaneous intestinal adenomas in the Apc Min mouse model by the nosteroidal anti-inflammatory drug piroxicam. . Cancer Res 56:710-4. Jemal A, Thomas A, Murray T, Thun M (2002) Cancer statistics. . Cancer J Clin 52:23- 47. . Jen J, Powell SM, Papadopoulos N, Smith KJ, Hamilton SR, Vogelstein B, Kinzler KW (1994) Molecular determinants of dysplasia in colorectal lesions. . Cancer Res 54(21):5523-6. Jenkins DJ, Wolever TM, Taylor RH (1981) Glycemie index of foods: a physiological basis for carbohydrate exchange. . Am J Clin Nutr 34:362—6. J in Z, Tamura G, Tsuchiya T. (2001) Adenomatous polyposis coli (APC) gene promoter hypermethylation in primary breast cancers. Br J Cancer 85: 69—73. J in Z, MacDonald RS (2002) Soy isoflavones increase latency of spontaneous mammary tumors in mice. J Nutr 132(10):3186-90. Jones J and Clemmons D (1995) Insulin-like growth factors and their binding proteins: biological actions. Endocr. Rev. 16:3-34. Kapila YL, Wang S, Dazin P, Tafolla 8, Mass MJ (2002) The heparin-binding domain and V region of fibronectin regulate apoptosis by suppression of p53 and c-myc in human primary cells. J Biol Chem 277(10):8482-91. Kashiwaba M, Tamura G, Ishida M (1994) Aberrations of the APC gene in primary breast carcinoma. J Cancer Res Clin Oncol 120:727—3 1. 198 Ke H, Pei J, Ni Z, Xia H, Qi H, Woods T, Kelekar A, Tao W (2004) Putative tumor suppressor LatsZ induces apoptosis through downregulation of Bel-2 and 8cl-x(L). Exp Cell Res 298(2):329-38. Kemler R (1993) From cadherins to catenins: cytoplasmic protein interactions and regulation of cell adhesion. . Trends Genet 9:317. Kennedy AR (1995) The evidence for soybean products as cancer preventive agents J Nutr, 125: 733S-743S. Kinoshita M, Kumar S, Mizoguchi A, Ide C, Kinoshita A, Haraguchi T, Hiraoka Y, Noda M (1997) Nedd5, a mammalian septin, is a novel cytoskeletal component interacting with actin-based structures. Genes Dev 11(12):1535-47. Kinzler KW, Vogelstein 8 (1996) Lessons from hereditary colorectal cancer. . Cell 87:159-70. Kirman I, Poltoratskaia N, Sylla P, Whelan RL (2004) Insulin-like growth factor-binding protein 3 inhibits growth of experimental colocarcinoma. Surgery 136(2):205-9. Kleeff J, Huang Y, Mok SC (2002) Down-regulation of DOC-2 in colorectal cancer points to its role as a tumor suppressor in this malignancy. Dis Colon Rectum 45: 1242—8. Kobayashi D, Yamada M, Kamagata C, Kaneko R, Tsuji N, Nakamura M, Yagihashi A, Watanabe N (2002) Overexpression of early grth response-1 as a metastasis-regulatory ' factor in gastric cancer. Anticancer Res 22(6C):3963-70. Koenuma M, Yamori T, Tsuruo T (1989) Insulin and insulin-like growth factor 1 stimulate proliferation of metastatic variants of colon carcinoma 26. Jpn J Cancer Res 80(1):51-8. Koivisto V, Yki-Jarvinen H (1993) Fructose and insulin sensitivity in patients with type II diabetes. . J Intern Med 233:145—53. Kristiansen E, Thorup IM (1995) Influence of different diets on development of DMH- induced aberrant crypt foci and colon tumor incidence in Wistar rats. Nutr Cancer. 1995;23(2):151-9. La P, Silva AC, Hou Z, Wang H, Schnepp RW, Yan N, Shi Y, Hua X (2004) Direct binding of DNA by tumor suppressor menin. J Biol Chem 279(47):49045-54. La Vecchia C, Negri E, Decarli A (1988) A case-control study of diet and colorectal cancer in northern Italy. Int J Cancer 41:492-98. La Vecchia C, Franceschi A, Dolora P, Bidoli E, Barbone F (1993) Refined sugar intake and the risk of colorectal cancer in humans. Int J Cancer 55:3 86-9. 199 La Vecchia C, Negri E, Decarli A, Franceschi S (1997) Diabetes mellitus and colorectal cancer risk. . Cancer Epidemiol Biomark Prev 6: 1007-101. Lahm H, c, Laurent PL, Fischer JR, Ceyhan A, Givel JC, Odartchenko N (1992) Growth regulation and co-stimulation of human colorectal cancer cell lines by insulin-like growth factor I, II and transforming growth factor alpha. Br J Cancer 65(3):341-6. Lamartiniere CA., Moore 1., Holland M. & 8ames S (1995) Neonatal genistein chemoprevents mammary cancer. Proc. Soc Exp Biol Med 208: 120-123. Lamartiniere CA, Moore J B, Brown NM, Thompson R, Hardin MJ & Barnes S (1995) Genistein suppresses mammary cancer in rats. Carcinogenesis 16:2833-40. Lasorella A, Noseda M, Beyna M, Yokota Y, Iavarone A (200) Id2 is a retinoblastoma protein target and mediates signalling by Myc oncoproteins. Nature 407:592-8. Lasorella A, Uo T, Iavarone A (2001) 1d proteins at the cross-road of development and cancer. Oncogene 20:8326-33. Lasorella A, Boldrini R, Dominici C (2002) Id2 is critical for cellular proliferation and is the oncogenic effector of N-myc in human neuroblastoma. Cancer Res 62:301-6. Le Marchand L, Wilkens LR, Kolonel LN, Hankin JH, Lyu LC (1997) Associations of sedentary lifestyle, obesity, smoking, alcohol use, and diabetes with the risk of colorectal cancer. . Cancer Res 57(21):4787-94. Lecuit T. (2005) Cell Adhesion: Sorting out Cell Mixing with Eehinoid? Curr Biol 15(13):R505-7. Lipkin M (1974) Phase I and phase II proliferative lesions of colonic epithelial cells in diseases leading to colonic cancer. . Cancer 34:878-88. Lipkin M, Blattner WA, Gardner EJ, Burt RW, Lynch H, Deschner E, Winawer S, Fraumeni J F Jr. (1984) Classification and risk assessment of individuals with familial polyposis, Gardner's syndrome, and familial non-polyposis colon cancer from [3 H]thymidine labeling patterns in colonic epithelial cells. Cancer Res 44(9):4201-7. Luceri C, Cademi G, Lancioni L, Aiolli S, Dolara P, Mastrandrea V, Seardazza F, Morozzi G (1996) Effects of repeated boluses of sucrose on proliferation and on AOM- induced aberrant crypt foci in rat colon. . Nutr Cancer 25: 1 87-196. Ma J, Pollak MN, Giovannucci 8, Chan JM, Tao Y, Hennekens CH (1999) Prospective study of colorectal cancer risk in men and plasma levels of insulin-like growth factor (IGF)-I and IGF-binding protein-3. . J Natl Cancer Inst 91 :620—5. 200 Macquart Moulin G, Riboli E, Cornee j, Kaaks R, Berthezene P (1986) A case-control study on colorectal cancer and diet in Marseilles. Int J Cancer 38: 183-191. Macquart Moulin G, Riboli E, Cornee j, Kaaks R, Berthezene P (1987) Colorectal polyps and diet: a case-control study in Marseilles. Int J Cancer 40: 179-188. Manousos 0, Day NE, Trichopoulos D (1983) Diet and colorectal cancer: A case-control study in Greece. Int J Cancer 32: 1-5. Martinez F, Rizza RA, Romero C (1994) High-fructose feeding elicits insulin resistance, hyperinsulinism and hypertension in normal dogs. . Hypertension 23:456—63. Mann 8, Gelos M, Siedow A, Hanski ML, Gratchev A, Ilyas M, Bodmer WF, Moyer MP, Riecken EO, Buhr HJ, Hanski C (1999) Target genes of beta-catenin-T cell- factor/lymphoid-enhancer-factor signaling in human colorectal carcinomas. Proc Natl Acad Sci U S A 16;96(4):1603-8. McGuinness 0P, Cherrington AD (2003) Effects of fructose on hepatic glucose metabolism.Curr Opin Clin Nutr Metab Care 6(4):441-8. McIntosh 0H, Regester 00, Le Leu RK, Royle PJ, Smithers GW (1995) Dairy proteins protect against dimethylhydrazine-induced intestinal cancers in rats. . J Nutr 125(4):809- l6. McKeown—Eyssen GE (1994) Epidemiology of colorectal cancer revisited: are serum triglyceride and/or plasma glucose associated with risk? Cancer Epidemiol Biomark Prev 3:687-95. Messina MJ, Persky V, Setchell KDR, 8ames S (1994) Soy intake and cancer risk: A review of in vitro and in vivo data. . Nutr Cancer 21 :1 13-31. Mocellin S, Provenzano M, Lise M, Nitti D, Rossi CR (2003) Increased TIA-1 gene expression in the tumor microenvironment after locoregional administration of tumor necrosis factor-alpha to patients with soft tissue limb sarcoma. Int J Cancer 107(2):317- 22. Mok SC, Chan WY, Wong K. DOC-2, a candidate tumor suppressor gene in human epithelial ovarian cancer. Oncogene 16:2381—7. Mi J, Kupfer GM. (2005) The F anconi anemia core complex associates with chromatin during S phase. Blood 105(2):759-66. Miller AB, Howe GR, Jain M (1983) Food items and food groups as risk factors in a case-control study of diet and colorectal cancer . Int J Cancer 32: 155-161. 201 Mills SJ, Mathers JC, Chapman PD, Burn J, Gunn A (2001 ) Colonic crypt cell proliferation state assessed by whole crypt microdissection in sporadic neoplasia and familial adenomatous polyposis. Gut 48(1):41-6. Moser AR, Dove WF, Roth KA, Gordon J I (1992) The Min (multiple intestinal neoplasia) mutation: its effect on gut epithelial cell differentiation and interaction with a modifier system. J Cell 8101.] 16(6): 15 1 7-26. Moser AR, Mattes EM, Dove WF, Lindstrom MJ, Haag JD, Gould MN (1993) APCMin, a mutation in the murine Ape gene, predisposes to mammary carcinomas and focal alveolar hyperplasias. Proc Natl Acad Sci U S A 90:8977—81. Murphy LJ, Friesen H0 (1988) Differential effects of estrogen and growth hormone on uterine and hepatic insulin-like growth factor I gene expression in the ovariectomized hypOphysectomized rat. Endocrinology 122:325—332. Murrill W8, Brown NM, Zhang JX, Manzolillo PA, Barnes S & Lamartiniere CA (1996) Prepubertal genistein exposure suppresses mammary cancer and enhances gland differentiation in rats. Carcinogenesis 17(7): 145 1-7. Muto T, Bussey HJ, Morson BC (1975) The evolution of cancer of the colon and rectum. Cancer 36(6):2251-70. Nishisho I, Nakamura Y, Miyoshi Y, Miki Y, Ando H, Horii A, Koyama K, Utsunomiya J, Baba S, Hedge P (1991) Mutations of chromosome 5q21 genes in FAP and colorectal cancer patients. . Science 253(5020):665-9. Nucci MR, Robinson CR, Longo P, Campbell P, Hamilton SR (1997) Phenotypic and genotypic characteristics of aberrant crypt foci in human colorectal mucosa. . Hum Pathol 28: 1396—407. Okazaki Y, F uruno M, F ANTOM Consortium, RIKEN Genome Exploration Research Group Phase I & 11 Team (2002) Analysis of the mouse transcriptome based on functional annotation of 60,770 full-length cDNAs. Nature 420(6915):563-73. Ooi GT, Tseng LY, Tran MQ, Rechler MM (1992) Insulin rapidly decreases insulin-like growth factor binding protein-1 gene transcription in streptozotoein-diabetic rats. . Mol Endocrinol 612219-28. Oshima M, Dinchuk J8, Kargman SL, Oshima H, Hancock 8, Kwong 8, Trzaskos JM, Evans JF and Taketo MM (1996) Supression of intestinal polyposis in APCA716 knockout mice by inhibition of cyclooxygenase-Z. . Cell 87: 803-9. Pagliassotti MJ, Prach PA (1995) Quantity of sucrose alters the tissue pattern and time course of insulin resistance in young rats. . Am J Physiol 269:R641-6. 202 Pamies-Andreu 8, F iksen-Olsen M, Rizza RA, Romero J C (1995) High-fructose feeding elicits insulin resistance without hypertension in nonna] mongrel dogs. . Am J Hypertens 8:732—8. Pasqualini R, Bourdoulous S, Koivunen 8, Woods VL Jr, Ruoslahti 8A (1996)Polymeric form of fibroneetin has antimetastatic effects against multiple tumor types. Nat Med 2:1197-203. Penhoat A, Naville D, Jaillard C, Chatelain PG, Saez JM (1989) Hormonal regulation of insulin-like growth factor I secretion by bovine adrenal cells. J Biol Chem 264:6858—62. Peters RK, Pike MC, Garabrant D, Mack TM (1992) Diet and colon cancer in Los Angels County, California. Cancer Causes and Control 3: 457-73. Peterson 0 & 8ames S (1991) Genistein inhibition of the growth of human breast cancer cells: independence from estrogen receptors and the multi-drug resistance gene. Biochem Biophys Res Commun 179:661-7. Peterson 0 & Barnes S (1996) Genistein inhibits both estrogen and growth factor- stimulated proliferation of human breast cancer cells. Cell Growth Differ 7:1345-51. Pickle LN, Greene MH, Ziegler RG (1984) Colorectal cancer in rural Nebraska. Cancer Res 44:363-369. Ponz de Leon M, Roncucci L, Di Donate P, Tassi L, Smerieri 0, Amorico MG, Malagoli G, De Maria D, Antonioli A, Chahin NJ (1988) Pattern of epithelial cell proliferation in colorectal mucosa of normal subjects and of patients with adenomatous polyps or cancer of the large bowel. Cancer Res 48(14):4121-6. Bleiberg H, Buyse M, Galand P (1985) Cell kinetic indicators of premalignant stages of colorectal cancer. Cancer 56(1): 124-9. Powell SM, 2112 N, Beazer-Barclay Y, Bryan TM, Hamilton SR, Thibodeau SN, Vogelstein B, Kinzler KW (1992) APC mutations occur early during colorectal tumorigenesis. . Nature 359:235-7. Pucci S (2004) Modulation of different clusterin isoforrns in human colon tumorigenesis, 23(13):2298-304. Poulsen M, Molck AM, Thorup I, Breinholt V, Meyer 0 (2001) The influence of simple sugars and starch given during pre- or post-initiation on aberrant crypt foci in rat colon. Cancer Lett 167(2): 135-43. Rajaram S, Baylink DJ, Mohan S (1997) Insulin-like growth factor-binding proteins in serum and other biological fluids: regulation and functions. Endocr Rev 18(6):801-31. 203 Rao CV, Wang CX, Simi B, Lubet R, Kelloff 0, Steele V, Reddy BS (1997) Enhancement of experimental colon cancer by genistein. Cancer Res 57(17):3717-22. Ratineau C, Bernard C, Poncet G, Blane M, Josso C, F ontaniere S, Calender A, Chayvialle JA, Zhang CX, Roche C (2004) Reduction of menin expression enhances cell proliferation and is tumorigenic in intestinal epithelial cells. J Biol Chem 279(23):24477- 84. Rebrin K, Steil GM, Getty L, Bergman RN (1995) Free fatty acid as a link in the regulation of hepatic glucose output by peripheral insulin. . Diabetes 44:1038—45. Reeves PG, Nielsen FH, Fahey GC (1993) AlN-93 purified diets for laboratory rodents: Final report of the American Institution of Nutrition Ad Hoc writing committee on the reformulation of the AIN-76A rodent diet. . J Nutr 123:1939-51. Reiser S, Handler H8, Gardner LB, Hallfrisch JG, Michaelis OE, Prather ES (1979) Isocalorie exchange of dietary starch and sucrose in humans: Effect on fasting blood insulin glucose, and glucagon and on insulin and gluccose response to a sucrose load. . Am J Clin Nutr 32:2206—16. Reiser S, Bohn E, Hallfrisch J, Michaelis 08 IV, Keeney M, Prather ES (1981) Serum insulin and glucose in hyperinsulinemic subjects fed three different levels of sucrose. . Am J Clin Nutr 34:2348-58. Russo MW, Sevetson BR, Milbrandt J (1995) Identification ofNABl, a repressor of NGFI-A- and Krox20-mediated transcription. Proc Natl Acad Sci U S A 92(15):6873-7. Ruzinova MB, Benezra R (2003) Id proteins in development, cell cycle and cancer. Trends Cell Biol 13:410-8. Sakatani T, Kancda A, Iacobuzio-Donahue CA, Carter MG, de Boom Witzel S, Okano H, Ko MS, Ohlsson R, Longo DL, Feinberg AP (2005) Loss of imprinting of Igf2 alters intestinal maturation and tumorigenesis in mice. Science 25;307(5717):1976-8. Sasai K, Katayama H, Stenoien DL, F ujii S, Honda R, Kimura M, Okano Y, Tatsuka M, Suzuki F, Nigg EA, Eamshaw WC, Brinkley WR, Sen S (2004) Aurora-C kinase is a novel chromosomal passenger protein that can complement Aurora-B kinase function in mitotic cells. Cell Motil Cytoskeleton 59(4):249-63. Santell RC, Kieu N & Helferich WG (2000) Genistein inhibits growth of estrogen- independent human breast cancer cells in culture but not in athymic mice. J Nutr 130:1665-69. Seaglia N, Igal RA (2005) Stearoyl-COA desaturase is involved in the control of proliferation, anchorage-independent growth, and survival in human transformed cells. J Biol Chem 280(27):25339-49. 204 Schroer A, Schneider S, Ropers H, Nothwang H (1999) Cloning and characterization of UXT, a novel gene in human Xpl 1, which is widely and abundantly expressed in tumor tissue. Genomics 56(3):340-3. Shao ZM, Wu J, Shen ZZ & Barsky SH (1998) Genistein exerts multiple suppressive effects on human breast carcinoma cells. Cancer Res 5 8:4851-57. Shiff SJ, Qiao L, Tsai LL, Rigas 8 (1995) Sulindac sulfide, an asprin-like compound, inhibits proliferation, causes cell cycle quiescence, and induces apoptosis in HT-29 colon adenocarcinoma cells. . J Clin Invest 96:491-503. Shostak K0, Dmitrenko VV, Garifulin OM, Rozumenko VD, Khomenko 0V, Zozulya YA, Zehetner G, Kavsan VM (2003) Downregulation of putative tumor suppressor gene TSC-22 in human brain tumors. J Surg Oncol 82(1):57-64. Scrensen IK, Kristiansen E, Mortensen AJ, Nicolaisen GM, Wijnands IAH, Kranen HJ Kreijl CF (1998) The effect of soy isoflavones on the development of intestinal neoplasia in Ape Min mouse. . Cancer Lett 130:217-25. Stamp d, Zhang XM, Medlline A, Bruce WR and C.Archer M (1993) Sucrose enhancement of the early steps of colon carcinogenesis in mice. Carcinogenesis, 14(4):777-9. Steil GM, Rebrin K, Mittelman SD, Bergman RN (1998) Role of portal insulin delivery in the disappearance of intravenous glucose and assessment of insulin sensitivity. . Diabetes 47:714—20. Stemmerrnan GN, Nomura AMY and Heilbrun LK (1984) Dietary fat and the risk of colorectal cancer. Cancer Res 44: 4633-7. Stephen AM, Haddad AC, Phillips SF (1983) Passage of carbohydrate into the colon: direct measurements in humans. . Gastroenterology 85(3):589-95. Stewart CE, Rotwein P (1996) Growth, differentiation, and survival: multiple physiological functions for insulin-like growth factors. Physiol Rev 76(4):]005-26. Stoffel W, Jenke 8, Block B, Zumbansen M, Koebke J. (2005) Neutral sphingomyelinase 2 (smpd3) in the control of postnatal growth and development. Proc Natl Acad Sci U S A 102(12):4554-9. Storlien LH, Kraegen 8W, Jenkins AB, Chisholm D (1988) Effects of sucrose vs starch diets on in vivo insulin action, therrnogenesis, obesity in rats. . Am J Clin Nutr 47:420-7. Strocchi A, Levitt M (1991) Measurement of starch absorption in humans. . Can J Physiol Pharmacol 69: 108-10. 205 Su LK, Kinzler KW, Vogelstein B, Preisinger AC, Moser AR, Luongo C, Gould KA, Dove WF (1992) Multiple intestinal neoplasia caused by a mutation in the murine homolog of the APC gene. . Science 256:668-70. Su LK, Vogelstein B, Kinzler KW (1993) Association of the APC tumor suppressor protein with catenins. . Science, 262: 1734-7. Takahashi Y, Miyoshi Y, Takahata C, Irahara N, Taguchi T, Tamaki Y, Noguchi S. (2005) Down-regulation of LATSl and LATSZ mRN A expression by promoter hypermethylation and its association with biologically aggressive phenotype in human breast cancers. Clin Cancer Res 11(4): 1 380-5. Taketo, MM (1998) Cyclooxygenase-Z inhibitors in tumorigenesis. . J Natl Cancer Inst 90: 1609-20. Tanaka TS, Jaradat SA, Lim MK, Kargul GJ, Wang X, Grahovac MJ, Pantano S, Sano Y, Piao Y, Nagaraja R, Doi H, Wood WH 3rd, Becker K0, K0 MS (2000) Genome-wide expression profiling of mid-gestation placenta and embryo using a 15,000 mouse developmental cDNA microarray. Proc Natl Acad Sci U S A 97(16):9127-32. Terpstra 0T, van Blankenstein M, Dees J, Eilers GA (1987) Abnormal pattern of cell proliferation in the entire colonic mucosa of patients with colon adenoma or cancer. Gastroenterology. 1987 Mar;92(3):704-8. Thiagarajan D, Bennink MR, Bourquin LD, Kavas FA (1998) Prevention of precancerous colonic lesions in rats by soy flakes, soy flour, genistein, and calcium. . Am J Clin Nutr 68:13948-98. ' Thiagarajan D, Bennink MR, Bourquin LD, Mayle JE, Seymour EM, Mridvika (1999) Effect of soy protein consumption on colonic cell proliferation in humans F ASEB J l3:A370. Thompson A, Morris R, Wallace M, Wyllie A, Steel C, Carter D (1993) Allele loss from 5q21 (APC/MCC) and l8q21 (DCC) and DCC mRNA expression in breast cancer. Br J Cancer 68:64—8. Thorling EB, Jacobsen N0, Overvad K (1993) Effect of exercise on intestinal tumor development in the male Fischer rat after exposure to azoxymethane. . Eur J Cancer Prev 2:77-82. . Thorbum AW, Storlien LH, Jenkins A8, Khouri S, Kraegen 8W (1989) F ruetose- induced in vivo insulin resistance and elevated plasma triglyceride levels in rats. . Am J Clin Nutr 49:1155—63. 206 Thresher J S, Podolin DA, Wei Y, Mazzeo RS, Pagliassotti MJ (2000) Comparison of the effects of sucrose and fructose on insulin action and glucose tolerance. . Am J Physiol 279:R1334-40 Thun MJ, Namboodiri MM, Heath CW (1991) Aspirin use and reduced risk of fatal colon cancer. . N Engl J Med 325 :1593-6. Tran TT, Medline A, Bruce WR (1996) Insulin promotion of colon tumors in rats. Cancer Epidemiol Biomarkers Prev 5( 12): 1013-5. Tricoli JV, Rall LB, Karakousis CP, Herrera L, Petrelli NJ, Bell 01, Shows TB (1986) Enhanced levels of insulin-like growth factor messenger RNA in human colon carcinomas and liposarcomas. Cancer Res 46:6169—73. Trougakos IP (2004) Silencing expression of the clusterin/apolipoprotein j gene in human cancer cells using small interfering RNA induces spontaneous apOptosis, reduced growth ability, and cell sensitization to genotoxic and oxidative stress. Cancer Res 64(5): 1 834- 42. Tsuda H, Sekine K, F uj ita K, Ligo M (2002) Cancer prevention by bovine lactoferrin and underlying mechanisms--a review of experimental and clinical studies. Biochem Cell Biol80(1):131-6. Tuyns AJ, Kaaks R, Haelterman M (1988) Colorectal cancer and the consumption of foods: a case-control study in Belgium Nutr Cancer 11:189-204. Underwood L E, Thissen J P, Lemozy S, Ketelslegers JM, Clemmons DR. (1994) Hormonal and nutritional regulation of IGF-I and its binding proteins. Horm Res 42: 145- 51. Valentinis 8, Bhala A, DeAngelis T, Baserga R, Cohen P (1995)The human insulin-like growth factor (IGF) binding protein-3 inhibits the growth of fibroblasts with a targeted disruption of the IGF-I receptor gene. Mol Endocrinol 9(3):361-7. Virkamaki A, Korsheninnikova E, Seppfiléi—Lindroos A (2001) Intramyocellular lipid is associated with resistance to in vivo insulin actions on glucose uptake, antilipolysis, and early insulin signaling pathways in human skeletal muscle. . Diabetes 50:2337-43. Wang Y, Li X, Wang L, Ding P, Zhang Y, Han W, Ma D (2004) An alternative form of paraptosis-like cell death, triggered by TAJ/TROY and enhanced by PDCDS overexpression. Journal of Cell Science 1 17: 1525-32. Wang Y, Putnam CD, Kane MF, Zhang W, Edelmann L, Russell R, Carrion DV, Chin L, Kucherlapati R, Kolodner RD, Edelmann W.(2005) Mutation in Rpal results in defective DNA double-strand break repair, chromosomal instability and cancer in mice. Nature Genetics 37:750-55. 207 Watanabe, Y, Tada, M, Kawamoto, K, Uozumi, G, Kajiwara, Y (1984) A case-control study of cancer of the rectum and the colon. . Nippon Shokakibyo Gakkai Zasshi 81: 185- 94. Weinberg 8D (2001) Human lactoferrin: a novel therapeutic with broad spectrum potential. Human lactoferrin: a novel therapeutic with broad spectrum potential. J Phann Pharmacol 53(10):]303-10. Weinberg RA (1989) Oncogenes, antioncogenes, and the molecular bases of multistepcarcinogenesis. . Cancer Res 49:3713-21. Weyer C, Funahashi T, Tanaka S (2001) Hypoadiponectinemia in obesity and type 2 diabetes: close association with insulin resistance and hyperinsulinemia. . J Clin Endocrinol Metab 86: 1930—5. Wiedmer T, Zhao J, Li L, Zhou Q, Hevener A, Olefsky JM, Curtiss LK, Sims PJ. (2004) Adiposity, dyslipidemia, and insulin resistance in mice with targeted deletion of phospholipid scramblase 3 (PLSCR3). Proc Natl Acad Sci U S A 101(36):]3296-301. Will JC, Galuska DA, Vinicor F, Calle 88 (1998) Colorectal cancer: another complication of diabetes mellitus? Am J Epidemiol 147:816-25. Wold MS. (1997) Replication protein A: a heterotrimeric, single-stranded DNA-binding protein required for eukaryotic DNA metabolism. Annu Rev Biochem 66, 61-92. Wolever TM, Jenkins DJ, Jenkins AL, Josse RG (1991) The glycemic index: methodology and clinical implications. . Am J Clin Nutr 54:846-54. Wolever TMS (2000) Dietary carbohydrates and insulin action in humans. . Br J Nutr 83(1):S97-S102. Wong JH, Ng T8. (2005) Sesquin, a potent defensin-like antimicrobial peptide from ground beans with inhibitory activities toward tumor cells and HIV-1 reverse transcriptase. Peptides 26(7):]120-6. Woodage T, King S, Wacholder S (1998) The APCII307K allele and cancer risk in a community-based study of Ashkenazi Jews. Nat Genet 20:62—5. Wu Y, Yakar S, Zhao L, Hennighausen L, LeRoith D (2002) Circulating insulin-like growth factor-I levels regulate colon cancer grth and metastasis.Cancer Res 62(4):]030-5. Wyllie AH (1985) The biology of cell death in tumours. . Anticancer Res 5(1):]31-6. 208 Xiong Z, Wu AH, Bender CM, Tsao JL, Blake C, Shibata D, Jones PA, Yu MC, Ross RK, Laird PW. (2001) Mismatch repair deficiency and CpG island hypermethylation in Sporadic colon adenocarcinomas. Cancer Epidemiol Biomarkers Prev 10(7):799-803. Yakar S, Liu JL, Stannard 8, Butler A, Accili D, Sauer 8, LeRoith D (1999) Normal growth and development in the absence of hepatic insulin-like growth factor-I. Proc Natl Acad Sci USA 96:7324—732. Yamada T, Suzuki M, Satoh H, Kihara—Negishi F, Nakano H, Oikawa T (2004) Exp Cell Res 294(1):39-50. Yang K, Fan K, Kurihara N, Shinozaki H, Rigas B, Augenlieht L, Kopelovich L, Edelmann W, Kucherlapati R, Lipkin M (2003) Regional response leading to tumorigenesis after sulindac in small and large intestine of mice with Ape mutations. . Carcinogenesis 24(3):605-1 1. Y1 M, and Ruoslahti E (2001) A fibronectin fragment inhibits tumor growth, angiogenesis, and metastasis. Proc Natl Acad Sci USA 98:620-4. Young D, Waitches G, Birchmeier C, F asano 0, Wigler M (1986) Isolation and characterization of a new cellular oncogene encoding a protein with multiple potential transmembrane domains. Cell 45(5):711-9. Yu H, Rohan T (2000) Role of the insulin-like grth factor family in cancer development and progression. JNCI 92(18): 1472-89. Zavaroni I, Chen YD, Reaven GM. (1982) Studies of the mechanism of fructose-induced hypertriglyceridemia in the rat. Metabolism 31(11):]077—83. Zenilman ME, Graham W (1997) Insulin-like grth factor-I mRNA in the colon is unchanged during neoplasia. Cancer Invest 15: 1-7. Zhang T, Nanney LB, Luongo C, Lamps L, Heppner KJ, DuBois RN, Beauchamp RD (1997) Concurrent overexpression of cyclin D1 and cyclin-dependent kinase 4 (Cdk4) in intestinal adenomas fi'orn multiple intestinal neoplasia (Min) mice and human familial adenomatous polyposis patients. . Cancer Res 57(1): 169-75. 209 I"[lli‘ljjljlljlljjjllljjlgljll