LIBRARY Michigan State University 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 AUG 2 8 2004 6/01 cJClRC/DateDuepGS-p. 1 5 INHIBITION OF INTESTINAL TUMORIGENESIS IN APCMIN MICE BY TART CHERRY ANTHOCYANINS AND SULINDAC BY Soo-Young Kang 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 Institute of Environmental Toxicology 2002 ABSTRACT INHIBITION OF INTESTINAL TUMORIGENESIS 1N ABCMIN MICE BY TART CHERRY ANTHOCYANINS AND SULINDAC By Soo-Young Kang Anthocyanins, a member of the flavonoid family of phytochemicals, previously have been found to inhibit the activities of cyclooxygenase (COX) enzymes. We hypothesized that tart cherry anthocyanins suppress intestinal tumorigenesis in ApcMin mice because compounds that inhibit COX potentially are protective against colon cancer development. A series of experiments were conducted to assess the influence of anthocyanins and a nonsteroidal anti- inflammatory drug, sulindac, on intestinal tumor development in ApcMin mice. First, we examined time frame of intestinal adenoma development in ApcMi" mice. The numbers and size of adenomas in small intestine increased with age, whereas tumor number in the cecum and colon did not increase significantly with age. The volume of colonic tumors increased significantly afier 75 days of age. These results characterized the early and rapid development and progression of intestinal tumors in ApcMi“ mice. We then conducted experiments to test the potential of anthocyanins to inhibit intestinal tumor development in ApcMin mice and growth of human colon cancer cell lines. Mice consuming anthocyanins and cyanidin in drinking water and a 20% tart cherry diet had fewer and smaller tumors in the cecum, whereas mice consuming sulindac in drinking water had more cecal adenomas. The mean diameter of adenomas in the small intestine was smaller in mice consuming sulindac, and larger in mice consuming the tart cherry diet, compared to mice consuming the control diet. Anthocyanins and cyanidin also reduced the growth of human colon cancer cell lines HT 29 and HCT 116. Based on these results, a second feeding study was conducted to determine effects of dietary anthocyanins and sulindac on 1) Min intestinal tumorigenesis in Apc mice, 2) expression and cellular localization of B- catenin, and 3) expression of target genes of the Wnt/B-catenin signaling pathway and global gene expression profiles. APCMin mice consuming dietary anthocyanins had significantly fewer adenomas in all three intestinal sections compared to mice consuming the control diet. Small intestinal adenomas in mice consuming sulindac were fewer in number and smaller in diameter compared to mice consuming anthocyanins, but mice consuming sulindac had more cecal adenomas compared to mice consuming anthocyanins. Sulindac did not influence the expression of B- catenin protein in intestinal epithelium, whereas anthocyanins increased B-catenin expression in the small intestine and cecum. Expression of mRN A of Apc, B-catenin and most of the target genes of Wnt/B-catenin signaling was not altered by treatment. cDNA microarray analysis identified several genes that were differentially expressed in response to treatment with anthocyanins or sulindac. Based on cellular expression of B-catenin and cDNA microarray analysis, we concluded that tumor inhibition in ApcMi" mice by anthocyanins is unlikely to involve COX inhibition or inhibition of the Wnt/B-catenin signaling pathway. Taken together, these results suggest that tart cherry anthocyanins and their aglycone, cyanidin, may reduce the risk of colon cancer by mechanisms that are unidentified at this time. T 0 my family iv ACKNOWLEDGEMENTS I would like to thank Dr. Les Bourquin for his guidance throughout the program as a major advisor. I appreciate his time and special effort put towards my research. I would also like to extend my gratitude to Dr. Maurie Bennink for his precious advice and kindness. Special thanks extend to Drs. Dale Romsos and Norm Hord for serving as the guidance committee members. And I am truly grateful for Dr. Steve Bursian for his support. He was always with me for the highs and lows and encouraged me to move on especially in many tough times of discouragement and depression. I thank Drs. Muralee Nair and Navindra Seeram for providing tart chenies and compounds for the experiments and their valuable suggestions during the lab meetings. Dr. John LaPres deserves sincere thanks for allowing me to use his lab and expertise for the microarray experiments. He has provided great help and support when I needed his advice. I would like to express my gratitude to Bing Wang, Elizabeth Rondini, Elizabeth Gignac and Crystal Ybarra for helping me with mice care and sacrifice. My sincere appreciation is extended to friends from the department. Drs. Mridvika, Anahita Mistry, Miyoung Jang, Sang-J in Chung and Joowon Lee have been source of cheer and advice throughout the program. I appreciate their friendship and advice. Thanks to Megan Waynar for her friendship and all those good times during the stressful days of exams and experiments. There were times that I thought I was walking in the tunnel of darkness and there were always my parents, Cheonun Kang and Gunja Moon, who did listen and pray for me to step forward. I thank them for their pride and faith in me. I also want to thank my late father-in-law for watching us up in heaven and mother-in-law for taking good care of Brandon and Chloe during my comprehensive and defense. My sisters-in-law, especially J inhee and Sunhee were great help on my busy days and I am thankful for their love on my two kids and compassion inherited from my mother-in-law. For my loving husband, J ae-ug, no words can easily describe how much I appreciate him for his love, affection and trust on me. I couldn’t have been through it without his limitless support, compliments and humor. At last, my two kids, Brandon and Chloe were always making me feel truly blessed and strong enough to shelter them and to face myself with no shame and fear. vi TABLE OF CONTENTS LIST OF TABLES ............................................................................................................. ix LIST OF FIGURES ........................................................................................................... xi CHAPTER I. INTRODUCTION ........................................................................................ 1 CHAPTER H. REVIEW OF LITERATURE ...................................................................... 6 l. Colon cancer .................................................................................................................... 7 2. The multi-step carcinogenic process ............................................................................... 8 3. Phytochemicals as anti-carcinogenic factors .................................................................. 9 4. ApcMm mouse model ..................................................................................................... 17 5. Nonsteroidal anti-inflammatory drugs (N SAIDs) and colorectal cancer ...................... 20 6. Genetics of intestinal neoplasia: APC mutation and its consequences ......................... 23 7. Hypotheses and research objectives .............................................................................. 33 CHAPTER III. ONTOGENY OF INTESTINAL ADENOMA DEVELOPMENT IN APCMIN MICE ................................................................................................ 36 1. Abstract ......................................................................................................................... 37 2. Introduction ................................................................................................................... 38 3. Materials and methods .................................................................................................. 40 4. Results ........................................................................................................................... 42 5. Discussion ..................................................................................................................... 5 1 CHAPTER IV. TART CHERRY ANTHOCYANINS INHIBIT TUMOR DEVELOPMENT IN ARCMIN MICE AND REDUCE GROWTH OF HUMAN COLON CANCER CELLS .............................................................................................. 54 1. Abstract ......................................................................................................................... 55 2. Introduction ................................................................................................................... 56 3. Materials and methods .................................................................................................. 58 4. Results ........................................................................................................................... 63 5. Discussion ..................................................................................................................... 73 CHAPTER V. TART CHERRY ANTHOCYANIN S REDUCE INTESTINAL TUMORIGENESIS AND ALTER INTESTINAL EPITHELIAL CELL GENE vii EXPRESSION, BUT DO NOT INFLUENCE INTRACELLULAR DISTRIBUTION OF B-CATENIN IN INTESTINE OF APCMIN MICE ...................................................... 77 1. Abstract ..................................................................................................................... 78 2. Introduction ............................................................................................................... 8O 3. Materials and methods .............................................................................................. 83 4. Results ....................................................................................................................... 93 5 . Discussion .............................................................................................................. 13 1 CHAPTER VI. OVERALL SUMMARY AND CONCLUSIONS ................................ 147 APPENDICES ............................................................................................................... 154 LITERATURE CITED ................................................................................................... 179 viii TABLE TABLE TABLE TABLE TABLE TABLE TABLE TABLE TABLE TABLE TABLE TABLE TABLE TABLE TABLE TABLE TABLE TABLE TABLE TABLE TABLE TABLE LIST OF TABLES 1: Lengths of small intestine and colon of all mice ........................................... 44 2: Composition of the experimental diets ......................................................... 59 3: Average intensity scores of B-catenin expression in intestinal sections ..... 104 4: Expression of B- catenin and target genes of Wnt/B- catenin signaling 1n intestinal sections of ApcMm mice ................................................................ 109 5: Expression of 8 genes influenced by treatment both in small intestine and colon ...................................................................................................... 111 6: Expression of 12 genes influenced by treatment both in small intestine and cecum .................................................................................................... 1 12 7: Expression of 20 genes influenced by treatment both in cecum and colon 1 l3 8: List of genes up-regulated by anthocyanins vs control and sulindac in cecum ........................................................................................................... 1 15 9: List of genes down-regulated by anthocyanins vs control and sulindac in cecum ........................................................................................................... 118 10: List of genes up- -regulated by anthocyanins vs control and sulindac 1n colon ............................................................................................................. 120 11: List of genes down-regulated by anthocyanins vs control and sulindac in colon ......................................................................................................... 122 12: List of genes up-regulated by sulindac vs control in small intestine ........ 124 13: List of genes up-regulated by sulindac and anthocyanins in small intestine ........................................................................................................ 126 14: List of genes down-regulated by sulindac vs control in small intestine 127 15: List of genes down-regulated by sulindac and anthocyanins in small intestine ........................................................................................................ 129 A1: Carrier status, body weights, and tissue lengths of mice from experiment I ................................................................................................. 155 A2: Numbers and sizes of adenomas in ApcMin mice from experiment I ........ 158 A3: Weekly body weights (g) of ApcMin mice from the experiment 11 ........... 160 A4: Tissue lengths and numbers and sizes of adenomas in ApcMin mice from experiment II ....................................................................................... 162 A5: Sex, carrier status, and issue lengths of mice from experiment III .......... 164 A6: Weekly body weights (g) of mice from experiment III ........................... 168 A7: Numbers and sizes of adenomas in ApcMin mice from experiment III ..... 172 ix TABLE A8: Intensity scores of B-catenin expression in small intestine of mice from experiment III ............................................................................................... 175 TABLE A9: Intensity scores of B-catenin expression in colon of mice from experiment HI ............................................................................................... 177 TABLE A10: Intensity scores of B-catenin expression in cecum of mice from experiment 1]] ............................................................................................... 178 FIGURE FIGURE FIGURE FIGURE FIGURE FIGURE FIGURE FIGURE FIGURE FIGURE FIGURE FIGURE FIGURE FIGURE FIGURE FIGURE FIGURE FIGURE FIGURE FIGURE FIGURE LIST OF FIGURES 1: The chemical structures of flavonoids. ................................................... 12-13 2: Anthocyanins fiom tart cherries .................................................................. 14 3: A genetic model for colorectal tumorigenesis .............................................. 25 4: Schematic of the APC protein primary structure .......................................... 26 5: APC-and NSAID-mediated suppression of intestinal tumorigenesis ........... 32 6: Influence of age on body weights ................................................................. 43 7: Influence of age on adenoma numbers in proximal (white), middle (gray) and distal (hatched) segments of the small intestine and total (black) numbers in the small intestine ........................................................... 45 8: Influence of age on the average diameter of small intestinal adenomas ...... 46 9: Influence of age on the average adenoma numbers in the cecum (gray) and colon (black) ............................................................................................ 48 10: Influence of age on adenoma volume (mm3) in the cecum ....................... 49 11: Influence of age on adenoma volume (mm3) in the colon ......................... 50 12: Influence of anthocyanins, cyanidin, tart cherries and sulindac on body weights (g) of mice throughout the experimental period .............................. 65 13: Influence of anthocyanins, cyanidin, tart cherries and sulindac on adenoma numbers in cecum (gray) and colon (black) ................................... 66 14: Influence of anthocyanins, cyanidin, tart cherries and sulindac on adenoma volumes (mm3) in cecum (gray) and colon (black) ....................... 67 15: Influence of anthocyanins, cyanidin, tart cherries and sulindac on adenoma numbers in small intestine .............................................................. 68 16: Influence of anthocyanins, cyanidin, tart cherries and sulindac on average diameters (mm) of adenomas in small intestine ............................... 69 17: Influence of anthocyanins on growth of human cancer cells, HCT 116 (gray) and HT 29 (black) ............................................................................... 71 18: Influence of cyanidin on grth of human cancer cells, HCT 116 (gray) and HT 29 (black) ............................................................................... 72 19: Aminoallyl labeling loop of RNA for microarray analysis ........................ 90 20: Influence of treatment on body weights (g) of mice throughout the experimental period ....................................................................................... 95 21: Influence of anthocyanins and sulindac on adenoma number in cecum (gray) and colon (black) ........ ' ......................................................................... 96 xi FIGURE 22: Influence of anthocyanins and sulindac on total adenoma volumes (mm3) in cecum (gray) and colon (black) ...................................................... 97 FIGURE 23: Influence of anthocyanins and sulindac on adenoma number in small ' intestine. ......................................................................................................... 98 FIGURE 24: Influence of anthocyanins and sulindac on mean diameters (mm) of adenomas in small intestine ........................................................................... 99 FIGURE 25: Expression of B-catenin in mice consuming the control diet ................... 101 FIGURE 26: Expression of B—catenin in mice consuming sulindac .............................. 102 FIGURE 27: Expression of B-catenin in mice consuming anthocyanins ..................... 103 FIGURE 28: Numbers of genes influenced by any treatment (P<0.05) in intestinal sections of APCM‘" mice ............................... ’ ............................................... l 10 xii CHAPTER I INTRODUCTION Colon cancer is the second most common cause of cancer mortality and the fourth most common in incidence in the United States (American Cancer Society, 2000). It has been estimated that dietary factors may account for 29 - 41% of cancers at all sites and that 66 - 75% of colon cancer is preventable by diet (WCRF/AICR, 1998). Results from epidemiological studies have shown that consuming fruits and vegetables is associated with reduced incidences of various cancers including colon cancer. The inverse association between cancer and fruit and vegetable intake may be due to number of components, including micronutrients and phytochemicals, contained in fruits and vegetables. Phytochemicals are defined as the naturally occurring, non-nutritive substances present in plant foods (Caragay, 1992). Studies have found that approximately 14 classes of dietary phytochemicals are present in common foods. These include sulfides, phytates, flavonoids, glucarates, carotenoids, coumarins, mono-terpenes, tri-terpenes, lignans, phenolic acids, indoles, isothiocyanates, phthalides, and polyacetylenes. Food sources that are rich in flavonoids are often found to be protective against colon cancer in animal models and humans. Anthocyanins, members of the flavonoid family of phytochemicals, are widely distributed in plants including beans, fruits, and vegetables, suggesting that plant-based diets can provide considerable amounts of anthocyanins (Harbome and Grayer, 1988). Anthocyanins primarily occur in plants as glycosides. C yanidin is the major anthocyanin aglycone in cherries. Montmorency and BalatonTM cherries contain 0.40 and 0.80 mg/g, respectively, of anthocyanins (Wang et al., 1999a). These anthocyanins were later found to be antioxidants and inhibit cyclooxygenase (COX) enzymes (Wang et al., 1999b). Cyanidin was intermediate in efficacy for inhibition of COX-1 and COX-2 when compared with aspirin and other non-steroidal anti-inflammatory drugs (N SAIDs) such as flurbiprofen and ibuprofen. Compounds that inhibit COX potentially are protective against colon cancer development since it has been demonstrated that NSAIDs inhibit the growth of colon tumors in animal models and reduce the risk of colon cancer in humans (Giovannucci et al., 1995; Labayle et al., 1991). Since anthocyanins have COX-2 inhibiting activity, this suggests they may have anticarcinogenic effects in the intestine. The ApcMin mouse strain is a mutant mouse lineage predisposed to multiple intestinal neoplasia (Min). This phenotype is the consequence of a mutation in the murine homolog of the adenomatous polyposis coli (APC) gene (Su et al., 1992). The APC gene is also mutated in humans who develop sporadic colon cancer as well as persons with familial adenomatous polyposis (FAP), an autosomal dominantly inherited disease that predisposes to colorectal cancer. The primary phenotype of mice carrying this mutation is the development of multiple intestinal adenomas, which progress to adenocarcinomas of the intestine in older mice. Min is transmitted by affected mice to 50% of the progeny with an unbiased sex distribution, as is characteristic of a fully penetrant autosomal dominant trait (Moser, 1990). Research on the effects of NSAIDs sulindac and piroxicam on tumor development in ApcMin mice showed that they reduced the incidence of intestinal tumors (Boobol et al., 1996; Jacoby et al., 1996). Studies have shown that NSAIDs possessing COX-2 inhibitory activity inhibited small intestinal tumori genesis, but not necessarily colonic tumorigenesis, in ApcMin mice (McEntee et al., 1999). In most cases, colon carcinogenesis depends on mutation of the APC gene and this gene is considered a gatekeeper in the carcinogenic process (Kinzler and Vogelstein, 1996). A major function of the APC protein is to bind cellular B-catenin and facilitate its degradation (Rubinfeld et al., 1993). Most mutations in the human APC gene have been found to occur between codons 1286 and 1513, which is within a region encoding B-catenin binding sites (Nakamura, 1993). These mutations generally result in the production of a truncated APC protein that cannot facilitate the degradation of B-catenin, suggesting that mutation of APC is essential to the process of intestinal tumor development by causing a loss of regulation of B-catenin expression. B-catenin functions as a component of the E-cadherin adhesion complex and also as a transcriptional regulator in a signaling pathway by binding T cell factor (Tet) transcription factor (Morin et al., 1997). Aberrant accumulation of B-catenin can influence trancriptional activation and expression of target genes in this signaling pathway. A few target genes in this pathway such as peroxisome proliferator activated receptor 8 (PPARO), c-MYC, cyclin D1 (CLNDl) and matrilysin (matrix metalloproteinase 7) recently have been identified (He et al., 1999; He et al, 1998; Tetsu and McCormick, 1999; Crawford et al., 1999). Cherries, anthocyanins and sulindac may influence tumorigenesis in the intestinal tract by modulating expression of genes involved in the carcinogenesis. The purpose of this research is to determine the effects of tart cherry anthocyanins and the NSAID sulindac on the development of intestinal adenomas and on gene expression in intestine, cecum and colon of ApcM‘n mice. CHAPTER II LITERATURE REVIEW Coloréfancer Colon cancer is the third leading cause of cancer mortality (after lung and stomach cancers) worldwide, with mortality estimated at 492,000 in 2001 (IARC, 2001). In the United States, colon cancer is the second most common cause of cancer mortality (after lung cancer) and is the fourth most common form of cancer in terms of incidence (after breast, prostate, and lung cancers) (American Cancer Society, 2000). Five-year survival following the diagnosis of colon cancer is approximately 55% in the United States and the rest of the developed world (Potter et al., 1993). Numerous studies have characterized the pathogenesis of colorectal cancer as a multistage process that begins with hyperproliferation of colonic mucosal epithelial cells. The intestinal epithelium has a complex architecture and undergoes continuous turnover. Maintenance of crypt structures requires control mechanisms that balance the rates of cell proliferation and cell death. Failure of the control mechanisms results in disorganization of the tissue and abnormal tissue growth. Normally, epithelial proliferation takes place at the base of mucosal glands, called the crypts of Lieberkuhn (Wright and Irvin, 1982). Quiescent (G0) cells then migrate to the superficial or luminal zone of the crypt, where they undergo apoptosis (programmed cell death) (Potten, 1996; 1997). Cell kinetic studies of rectal mucosal biopsies determined that patients at increased risk of developing colorectal cancer have an expansion of the proliferative compartment and distribution of S-phase cells toward the lumenal zone of the crypt (Terpstra et al., 1987). Hyperproliferation may contribute to mutations that lead to the formation of adenomas, the precursor lesions to colonic carcinoma. To prevent development of colon cancer, it has been suggested that the multistage carcinogenesis process must be delayed or pre-empted. This could be achieved by modifying the diet to include more cancer preventive foods, since it has been estimated that 66 - 75% of colon cancer is preventable by adequate diets (WCRF/AICR, 1998). Indeed, results from epidemiological studies have shown that consuming diets rich in fruits and vegetables is associated with reduced incidences of various cancers including colon cancer (Willet, 2001). The Multi-Step Carcinogenic Process Carcinogenesis is a multistep process. Moolgavkar and Knudson (1981) suggested two distinct stages, preneoplastic and malignant steps. Miller and Miller (1981) summarized the carcinogenisis process as follows: “Initiation” is the cellular change forming the first preneoplastic (initiated) cell in a tissue. The chemically induced initial step from a normal cell to a premalignant cell has been shown to be irreversible in mouse skin (Berenblum and Shubik, 1947). “Promotion” can be defined as the action of any compound that increases the tumor size by expanding the pool of initiated cells. Treatment with promoting substances shortens the long latency of tumor development and the action of these substances is reversible (Miller and Miller, 1981). The irreversible processes of visible tumor formation thereafter can be termed “progression”. Progression can be considered the terminal stage of neoplastic development (Pitot, 1989). Phytochemicals as Anti-Carcinogenic Factors It has been estimated that dietary factors may account for 29 - 41% of cancers at all sites and that 66 - 75% of colon cancer is preventable by diet (WCRF/AICR, 1998). Several human cohort and case-control studies for a number of common cancer sites have shown that increased intakes of fruits and vegetables are associated with decreased cancer incidence and mortality rates (Doll, 1990; Ocke et al, 1997). The inverse association between cancer risk and fruit and vegetable intake may be due to a number of components, including micronutrients and phytochemicals, contained in fruits and vegetables. Phytochemicals are defined as the naturally occurring non—nutritive substances present in plant foods (Caragay, 1992). Studies have found that approximately 14 classes of dietary phytochemicals are present in common foods. These include sulfides, phytates, flavonoids, glucarates, carotenoids, coumarins, mono-terpenes, tri-terpenes, lignans, phenolic acids, indoles, isothiocyanates, phthalides, and polyacetylenes. Some of these components may potentially block metabolic pathways leading to carcinogenesis and have been found to decrease cancer risk in experimental animals. The effect of the substances may depend on animal species, structure of the carcinogen, dose regimen, timing of dosages, and many other experimental parameters. The effective doses for some anticarcinogens may exceed what humans ingest from plant sources. On the other hand, the combination of several anticarcinogens at sub-effective levels has been shown to result in antitumor effects in some cases (Renner, 1990). Food sources that are rich in flavonoids are often found to be protective against colon cancer. Approximately 4,000 different flavonoids have been identified in many plant families (Harbome, 1988). The flavonoid family includes a wide variety of chemical structures and are the largest group of plant phenolics (King and Young, 1999). The flavonoids (Fig. l) are polyphenolic compounds possessing 15 carbon atoms; two benzene rings joined by a linear three carbon chain. These low molecular weight compounds are usually glycosylated on one or more of their hydroxyl groups. Many flavonoids are easily recognized as flower pigments in most angiosperm families (flowering plants). However, their occurence is not restricted to flowers and in fact can be found in all parts of the plant. Major classes of flavonoids include flavanones, C-glycosylflavonoids, isoflavonoids, and anthocyanins (Fig. 1). Anthocyanins are widely distributed in plants including beans, fruits, and , vegetables (Harbome and Grayer, 1988). There are more than 240 known structures for anthocyanins based on 6 major aglycones-pelargonidin, cyanidin, peonidin, delphinidin, petunidin, and malvidin. Anthocyanins are universal plant colorants and largely responsible for colors of flower petals and fruits of higher plants (Harbome, 1988). Another function of anthocyanins includes light absorbance, thereby protecting the photosynthetic system from excess light (Timberlake, 1980). Cyanidin based anthocyanins have been found in a wide variety of berries and cherries as well as sweet potatoes, red onions and currants. Characterization of tart cherries (Wang et al., 1997) showed that the predominant anthocyanins in Balaton and Montmorency cherries were anthocyanin 1, anthocyanin 2 and anthocyanin 3 (Figure 2). Wang et al. (1999a) reported that 100 g of fresh and pitted cherries 10 contain 12.5 - 25.0 mg of anthocyanins. Anthocyanins 1 — 3 and the parent aglycone, cyanidin, were found to possess antioxidant and anti-inflammatory activities (Wang et al., 1999b). The antioxidant activities of anthocyanins were superior to that of or- tocopherol and comparable to that of the commercial antioxidants butylated hydroxyanisole (BHA) and butylated hydroxytoluene (BHT). C yanidin inhibited cyclooxygenase (COX) 1 and 2 enzymes with ICSO values of 90 and 60 11M, respectively (Wang et al., 1999b). Cyanidin was found to be four times more powerful as an antioxidant than Trolox, a vitamin E analogue (Rice-Evans, 1995). Tsuda et al. (1996) reported inhibition of lipid peroxidation and the active oxygen radical scavenging effect of anthocyanin pigments and cyanidin 3-O-beta-D-glucoside (C3G). Dietary cyanidin C3G also resulted in a significant decrease in generation of thiobarbituric acid reactive substances and showed a significantly lower susceptibility to lipid peroxidation in rats (Tsuda, 1988). Cyanidin has been found to function as a potent antioxidant in vivo in another recent study (Tsuda, 2000). C3G showed a potency to scavenge active oxygen species in rats fed a diet containing C3G (2 g/kg diet) for 14 days, and then subjected to hepatic ischemia-reperfusion (I/R) as an oxidative stress model (Tsuda, 2000). Ramirez-Tortosa et al. (2001) have shown that an anthocyanin-rich extract decreased lipid peroxidationand DNA damage in vitamin E-depleted rats. Seeram et a1 (2001) reported cyclooxygenase inhibitory and antioxidant effects of cyanidin glycosides from cherries and berries. The antioxidant activity of anthocyanins was comparable to commercial antioxidants and superior to 11 a) The skeleton structure of flavonoids b) Chalcones OH HO 0 c) Flavones OH HO I 0 H0 0 d) Flavonols OH HO 0 Figure 1. The chemical structures of flavonoids. 12 e) Flavanone OH HO 0 HO 0 f) Isoflavones HO o 0 Ho OH g) Anthocyanins OH HO 0 OH 110 Figure 1 continued. The chemical structures of flavonoids. OR2 OH Figure 2. Anthocyanins from tart cherries Anthocyanin l. R1=glucose, R2: rhamnose, [3-cyanin 2’ ’-O-B-D-glucopyranosyl-6’ ’-O-or-L-rhamnopyranosyl-B-D- glucopyranoside] Anthocyanin 2. R1=H, R2= rhamnose, [3-cyanin 6’ ’-O-or-L-rhamnopyranosyl-B-D-gluc0pyranoside] Anthocyanin 3. R1=R2=H, [3-cyanin O-B-D—glucopyranoside] l4 vitamin E whereas the COX inhibitory activities were comparable to those of ibuprofen and naproxen at 10 uM concentrations (Seeram et al, 2001). In general, anthocyanins are stable in acidic conditions (below pH 7) and unstable in alkaline conditions. Anthocyanins can be hydrated at the C-2 position and degraded to Chalcones in alkaline conditions of pH 7 or higher. A mechanistic study done by Tsuda et a1. (1996) showed that C3G had strong antioxidative activity even at neutral pH. These results indicate that the flavylium cation form, which predominates at acidic pH, may not be essential for the antioxidative activity of anthocyanins. The study also showed that C3G reacted with azo-compounds to generate alkylperoxyl radicals. The reaction products were identified as 4,6-dihydroxy-2-0-b-D-glucosyl- 3-oxo-2,3-dihydrobenzofuran and protocatechuic acid (Tsuda et al., 1996). Thus, degradation of anthocyanin pigments could produce additional radical scavengers. A subsequent study on absorption and metabolism of C3G (Tsuda et al., 1999) showed that C3G was rapidly found in the plasma in rats ingesting 0.9 mmol/kg body weight of C3G by direct stomach intubation, indicating that the glycoside can be absorbed intact. Protocatechuic acid was found in the plasma at a concentration 8-fold higher than that of C3G, whereas cyanidin was not detected in plasma (Tsuda et al., 1999). Miyazawa et a1. (1999) also found that oral supplementation of C3G and cyanine-3, 5-diglucode incorporated into plasma and liver of rats and human plasma by UV- HPLC. Little research has been conducted for the effect of anthocyanins and cyanidin on colon carcinogenesis. Koide et a1. (1996) first reported that feeding red rice prolonged survival of Balb/C mice inoculated i.p. with Meth/A lymphoma cells 15 and that hydrolyzed anthocyanins decreased the growth of HCT-1 5 cancer cells. Later, they found that sugar-bonded bioflavonoids extracted from red soybeans and red beans prolonged survival of Balb/C mice bearing tumor Meth/A cells and that anthocyanin fractions extracted fiom red soybeans inhibited the growth of HCT-1 5 cells (Koide et al., 1997). Pool-Zobol et al. (1999) have shown that anthocyanins reduced HzOz-induced DNA strand breaks but did not prevent endogenous generation of oxidized DNA bases in HT 29 clone 19A human colon cancer cells. Recently, Meiers et a1. (2001) investigated the influence of anthocyanins on the growth of human tumor cells. They reported that the aglycones of anthocyanins, cyanidin and delphinidin inhibited the growth of human vulva carcinoma cell line A431 and their ICso values were 42 and 18 uM, respectively. Cyanidin (0.8 i 0.2 uM ) and delphinidin (1.3 i 0.2 1.1M) were potent inhibitors of the tyrosine kinase activity of the epitherrnal growth-factor receptor (EGFR). Treatment with an irreversible inhibitor of the EGFR-kinase, EKI-569, in combination with sulindac showed remarkable protection from tumor development (95% reduction in polyp number) in ApcMin mice (Torrance et al., 2000). Recently, it has been reported that purple corn anthocyanins suppressed the promotion of colon tumors caused by 2- amino-l-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP) in rats initiated with dimethyl hydrazine. However, these researchers did not test the potential of purple corn color anthocyanins to directly suppress tumor development (Hagiwara et al., 2001). 16 ApcMi" Mouse Model The ApcMin (mutiple intestinal neoplasia) mouse has been proposed to be a usefirl model for the study of human colorectal cancer. Moser et a1. (1990) reported a mutation in the mouse genome that resulted in the development of multiple intestinal tumors in all carriers of the mutation. The mutation was identified in a pedigree established during a mutagenesis project in which C57BL/6J (B6) males were treated with ethylnitrosourea and then mated to AKR/J (AKR) females. The primary phenotype of ApcMin mice is the development of multiple adenomas that progress to adenocarcinomas of the intestine in older mice. Min is transmitted by affected mice to 50% of progeny with an unbiased sex distribution, as is characteristic of a fully penetrant autosomal dominant trait (Moser, 1990). The mutation is currently maintained by crossing Min/+ males with B6 females. Tumors usually are present throughout both the small and large intestine and the cecum, but are not present in the stomach. ApcMin mice are heterozygous for a nonsense mutation (ApcMin ) at the Apc locus, the murine homologue of the adenomatous polyposis coli (APC) gene (Su et al., 1992). Two candidate tumor suppressor genes, APC and MCC (mutated colon cancer) on human chromosome 5q21 frequently are mutated in sporadic colorectal cancers (Kinzler et al., 1991). The APC gene is also mutated in humans with familial adenomaous polyposis (F AP), an autosomal dominantly inherited disease that predisposes to colorectal cancer. The frequency of somatic APC mutations is the same in adenomas and carcinomas, suggesting that mutation of APC is an early 17 event in sporadic colorectal cancer (Levy et al., 1994). Using a site-specific quantitative polymerase chain reaction assay, Luongo et a1. (1994) found extensive loss of the remaining wild-type Apc (Apc+) allele at the Apc locus in spontaneously- occurring intestinal adenomas from mice heterozygous for the ApcMin nonsense mutation. Examination of human FAP kindreds demonstrates that family members who inherit the same APC mutation may differ dramatically in tumor burden (Leppert et al., 1990). Although environmental factors may be partially responsible, modifier genes have been proposed to account for some of this variability (Paul et al., 1993; Giardiello et al., 1994). Further study of Min-induced neoplasia by manipulation of the mouse gerrnline identified genes modifying the Min phenotype: Moml(modifier of Min 1), genes for the 5-cytosine DNA methyltransferase, and DNA mismatch repair factor Msh 2 (Bilger et al., 1996). Quantitative trait loci mapping identified the Mom 1 locus in the distal region of mouse chromosome 4 (McPhee et al., 1995). Mom 1 also resides in a region of synteny with human chromosome 1p35, a region frequently associated with loss of heterozygosity in neuroblastoma and colon cancer (Martinsson et al., 1989; Leister et al., 1990). The gene for secretory type II phospholipase A2 (sPLA2) maps to the same region that contains Mom 1 (McPhee et al., 1995). The sPLA2 are extracellular enzymes that, upon activation, cleave acyl ester bonds in the sn-2 position in glycerophospholipid. These enzymes play a crucial role in the generation of arachidonates and eicosanoids, proinflammatory mediators. 18 Therefore, the Min mutation exerts a decisive effect in a pathway leading to intestinal tumor formation. However, the fact that all cells of a ApcMin mouse carry this predisposing gerrnline alteration, but only a limited number of tumors develop, indicates that somatic events are also necessary for tumor formation. Min-dependent tumorigenesis also occurs with less frequency in mammary glands, the pancreas, and the body wall (Bilger et al., 1996). Mouse models such as the Min mutant should provide a major resource for studying the anticarcinogenic potential of dietary factors, pathways of tumorigenesis, and the molecular identification of genes that can control particular neoplastic processes. There has been great interest in studying the effects of dietary or other interventions on colon carcinogenesis. However, most research has been conducted using animal models of carcinogenesis with tumors initiated by injecting large doses of carcinogens and using different experimental conditions (i.e. different carcinogens, injection doses, etc). This makes it difficult to compare results from studies conducted in different laboratories. The ApcMin mouse has been used as a model to study effects of dietary factors on human colorectal cancer. Several studies have been conducted to study the potential of diet to inhibit the formation of intestinal tumors in ApcMin mice. Bowman-Birk protease inhibitor concentrate (0.5%) from soybeans reduced colonic tumors and small intestinal tumors in Min mice by approximately 40% (Kennedy et al., 1996). Paulsen et a1 (1997) showed that eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA)- enriched fish oil suppressed tumor number and burdon in ApcMin mice. Rye-bran Min diet also has been shown to prevent the formation of intestinal polyps in Apc mice 19 (Mutanen et al., 2000). Mahmoud et a1. (2000) reported that plant phenolics, including caffeic acid phenethyl ester and curcumin, decreased intestinal tumor formation by 63% in ApcMin mice. Green tea extract and sulindac acted Mm mice synergistically to inhibit development of intestinal tumors in Apc (Suganuma et al., 2001). Treatment with both green tea extract and sulindac significantly reduced the number of tumors from 72 to 32 tumors whereas green tea extract or sulindac alone reduced tumor number to 57 or 49, respectively (Suganuma et al, 2001). Results from studies using ApcMin mice have not always supported the findings of epidemiological studies and studies with carcinogen-injected animal models. Some dietary factors do not influence tumor development when administered to ApcMin mice. Pierre et a1. (1997) reported that addition of short chain fructooligosaccharides to the diet reduced colonic tumor incidence but had no effect on small intestinal tumors in ApcMin mice. Van Kranen et a1. (1998) found no effect of dietary fat or a fruit and vegetable mixture on tumor formation in ApcMin mice. Sorenson et a1. (1998) reported that soy isoflavones had no effect on tumor Min formation in Apc mice. N onsteroidal Anti-Infammatory Drugs (NSAIDsland Colorectal Cancer Several lines of investigation have demonstrated that nonsteroidal anti- inflammatory drugs (N SAIDs) inhibit the growth of colon tumors in animal models and reduce the risk of colon cancer in humans. Giovannucci et a1. (1995) found that four to six tablets of aspirin per week sustained over years substantially reduced the 20 risk of colorectal cancer. Other NSAIDs such as sulindac and ibuprofen have been shown to reduce the relative risk of colorectal cancer afier 2 or more years of continuous use (Muscat et al., 1994). The mechanism of the anticarcinogenic effect of these drugs has not been fully established. One potential explanation for this protective effect is inhibition of intestinal epithelial growth. It has been demonstrated that NSAIDs, including indomethacin and sulindac, inhibit the rate of growth of carcinogen-induced colon tumors in animal models (Olsson et al., 1984; Skinner et al., 1991). Levi et al. (1990) showed that indomethacin inhibits the gastric epithelial cell hyperproliferation that normally restores mucosal integrity afier injury in rodent models. Studies of sulindac in FAP patients provide further evidence that NSAIDs inhibit epithelial cell proliferation in the gastrointestinal tract. Regression of colorectal polyps was observed after 6 months of treatment with oral sulindac (Labayle et al., 1991; Rigau et a1.; 1991). Polyps returned afier treatment was discontinued. Reductions of mucosal prostaglandin E2 levels paralleled polyp regression (Labayle et al., 1991). Most hypotheses for explaining the anticarcinogenic effects of NSAIDs have focused on the common property of NSAIDs to inhibit COX enzymes and thereby cause a subsequent reduction in levels of prostaglandins in tissue. Inducible COX-2 is overexpressed in human colon cancers. Sulindac inhibited tumor formation and reduced overexpression of COX-2 level in Min mice (Boolbol et al., 1996). Jacoby et al. (1996) demonstrated that dietary piroxicam significantly reduced intestinal tumor multiplicity in Min mice to 12% of that observed in control animals. Plasma 21 thromboxane B2 levels were reduced in parallel with tumor inhibition (Jacoby et al., 1996). Ritland and Gendler (1999) confirmed chemopreventive effects of proxicam Min on intestinal adenomas in Apc mice. Short-term administration of proxicam (200 Mi" mice with the ppm) reduced tumor multiplicity by 90%. Treatment of Apc NSAID nabumetone resulted in a dose dependent suppression of intestinal tumorigenesis (Roy et al., 2001). Nabumetone induced apoptosis by Min downregulating the anti-apoptotic protein, Bcl-2, in the intestine of Apc mice. McCracken et al. (1996) investigated the effect of COX-inhibiting (S-form) and non-COX-inhibiting (R-form) enantiomers of flurbiprofen on rat colonocyte proliferation. Sprague Dawley rats were treated orally with R- or S-flurbiprofen or vehicle. Both R and S forms significantly reduced colonocyte labeling index, by 34% and 23%, respectively, compared with vehicle. R-flurbiprofen caused minimal ulcer formation (4.48 mmz) compared with S-flurbiprofen (94.4 mmz). These findings suggest that R-flurbiprofen-mediated control of colonocyte proliferation is independent of prostaglandin biosynthesis. Shiff et al. (1995) showed a possible alternate mechanism by which NSAIDs inhibit gastrointestinal cell proliferation. They demonstrated that both sulindac and sulindac sulfide caused cell cycle quiescence and induced apoptosis by inhibition of the p33 cdk prOtein, a key regulator of the cell cycle. Sulindac was effective despite its relative weak COX- inhibitory activity compared to sulindac sulfide. Notably, disruption of the mouse sPLA2 locus has been shown to increase tumor number and restoring sPLA2 expression through transgenic constructs decreases tumor number in ApcM'n mice, 22 despite the prediction that sPLA2 should decrease prostaglandin production (Kennedy et al., 1995; Cormier et al., 1997). The clinical and experimental evidence that NSAIDs are anticarcinogenic is strong, and several studies have suggested COX-independent mechanisms for this action. The exact relationship of prostaglandin synthase inhibition by NSAIDs to tumor prevention remains uncertain. Genetics of Intestinal Neoplasia: APC Mutation and its consequences Different forms of human colon cancer share mutations in APC and may also share other genetic features (Fearon and Vogelstein, 1990). Certain mutations are common in colon cancer, but the sequence of events may vary. Temporal series of somatic changes including mutation of APC, K-RAS, DCC and P53 and hypomethylation of DNA have been demonstrated by molecular analysis of human intestinal cancer (Fig. 3). This observation shows that a particular molecular change is correlated with a particular stage in the carcinogenesis. It generates the hypothesis that certain gene mutations are necessary for particular carcinogenic steps in colon carcinogenesis. In most cases, colon carcinogenesis depends on mutation of the APC gene and it is considered a gatekeeper in the carcinogenic process (Kinzler and Vogelstein, 1996). APC encodes a cytoplasmic polypeptide of 2843 residues (Fig. 4). The N-terminus contains oligomerization sites indicating that the molecule acts as a polymer (Su et al., 1993a). The central part of the molecule contains sites for 20 amino acid [3- catenin binding repeats and also contains heptad repeats and armadillo (Arm) repeats 23 (Su et al., 1993b). The heptad repeats lead to supercoiling of helix and arm repeats composed of three alpha helices mediate protein-protein interactions. The truncations in the midpoint cluster region, which eliminate all the axin-binding and most of the B-catenin-binding repeats, account for more than 60% of oncogenic mutations in APC (Miyoshi et al., 1992). The C terminal region contains tubulin binding sites (Munemitsu et al., 1994; Smith et al., 1994). Tubulin is the protein that polymerizes into long chains or filaments that form microtubules, hollow fibers which serve as a skeletal system for processes such as cell division and the movement of materials within cells. The region also contains binding sites for the human homologue of the Dlg (Drosophila large discs) tumor supressor protein. 24 Chromosome Sq 12p 18q 17p Alteration _ Loss Activation Loss Loss Gene APC k-ras DCC p53 Hypomethylation V V V Normal Hyperproliferative + Adenoma Epithelium epithelium Early—-> lntennediate —> Late Y Carcinoma Figure 3. A genetic model for colorectal tumorigenesis (modified from Vogelstein and Kinzler, 1993) 25 Heptad repeats Three lS-amino acid repeats DLG binding sites ¢—> I | | I | l <———> ‘ ’ N D-D-Doooo-ooo C / |||||| , , Oligomerization ‘—> ¢ > Domain Arm repeats Seven 20-amino acid Tubulin binding sites B-catenin binding repeats <———> Midpoint Cluster Region Figure 4. Schematic of the APC protein primary structure (modified from Spink et al., 2001) 26 It has been shown that intestinal adenomas in ApcMin mice and human colon tumor cell lines have lost the wild type allele or express only short N-temrinal fragments of the Apc polypeptide (Luongo et al., 1994). The Min allele contains a nonsense codon at position 850 (Moser et al., 1990) and other targeted mutant Apc alleles have been developed that result in Apc polypeptide truncation of codons 716, 1638 and 474 by Oshima et a1. (1995), Fodde et a1. (1994), and Sasai et al. (2000), respectively. Ape“6 mice are predisposed to develop 200 - 500 adenomas mainly in the small intestine, whereas Apc1638 are predisposed to develop 5 - 6 adenomas and adenocarcinomas along the intestinal tract and a number of extra-intestinal lesions including mammary tumors (Oshima et al., 1995; Fodde et al., 1994). Apc474 mice develop around 100 intestinal polyps and 18% of them manifested mammary tumors (Sasai et al., 2000). The APC cytoplasmic polypeptide plays a regulatory part in the cell and participates in several protein assemblies. Most human APC mutations are found between codons 1286 and 1513 and this midpoint mutation cluster region occurs near the B-catenin binding sites (Nakamura, 1993). Mutations result in the production of a truncated protein which cannot facilitate the degradation of B-catenin, suggesting that mutated APC is essential in the process of tumor development by causing a loss of regulation of B-catenin. B-catenin performs different functions within the cell. Cytoplasmic and nuclear B—catenin accumulate only in response to activation of the Wnt/Wingless growth factor signaling pathway whereas a membrane associated pool is bound to E- 27 cadherins and or—catenin in the adherins junction (Yap etal., 1997). E-cadherin is one of a large family of genes which code for calcium-dependent cell adhesion molecules (Takeichi et al., 1988). Any free B-catenin not bound to E-cadherens is targeted for degradation in the absence of a Wnt signaling. The APC gene product is required in the B-catenin degradation process as an essential part of a mutiprotein complex. A complex of glycogen synthase kinase (GSK)-3B and conductin/axin phosphorylates the APC/B-catenin complex, which induces ubiquitination and degradation of B-catenin by proteosomes (Easwaran et al., 1999). The ubiquitination and degradation of B-catenin is a highly regulated process and requires a multi- molecular complex with APC, conductin/axin, and GSK-3B. APC mutation allows increased levels of cytoplasmic B-catenin, which may then trigger a cascade of events resulting in the initiation of adenomas. The cytoplasmic B-catenin is translocated to the nucleus and functions as a transcription factor in intracellular signaling. B-catenin binds to T cell factor(ch)/lymphoid enhancer factor (Let) transcription factors in the nucleus (Ilyas and Tomlinson, 1997). Thus, APC mutation causes aberrant accumulation of B-catenin, which then translocates to the nucleus and binds ch/Lef , causing increased transcriptional activation of some genes. These events ultimately result in abnormal cell growth since normal APC function is essential for maintaining normal cell architecture and growth. Genes that might be targets for transcriptional activation by B-catenin/ch transactivation have been identified. Crawford et al. (1999) suggested that matrilysin 28 is a target gene for B-catenin/ch transactivation. Matrilysin is a matrix metalloproteinase 7 (MMP~ 7) expressed in approximately 90% of human colonic adenocarcinomas and in many human colonic tumor cell lines (N ewell et al., 1994). Cyclin D1 (CLNDl) has also been identified as a target gene of B-catenin transactivation (Tetsu and McCormick, 1999). CLNDl is a regulator of the cell cycle and is highly expressed in adenomatous polyps and colorectal adenocarcinomas (Zhang et al., 1997). Overexpression of CLND 1 may derange the cell cycle machinery and contribute inappropriate neoplastic cell growth (Hartwell and Kastan, 1994). He et al. (1998) identified c-MYC as a target gene in the signaling pathway of APC mutation. C-MYC expression mediated through ch—4 binding sites in the promoter region is repressed by APC and activated by B-catenin. He et al. (1999) also reported that peroxisome proliferator activated receptor 6 (PPARO) is a target of the APC tumor suppressor pathway and is an APC-regulated target of NSAID drugs. They found that APC repressed PPAR5 expression and that PPARS activity was regulated by the APC/B-catenin/ch-4 pathway when APC was introduced in HT-29 cell lines. Interestingly, it has been suggested that PPARs may be a target of NSAIDs in suppressing tumorigenesis since NSAIDs can perturb eicosanoid metabolism and mediators involved in eicosanoid metabolism are known ligands for PPARs (Prescott and White, 1996). Sulindac sulfide resulted in a dose- dependent repression of PPARS activity in HCT116 and SW480 cell lines by disrupting the DNA binding ability of PPARO/RXROL (retinoid X receptor or) heterodimers (He et al., 1999). Recently, the role of PPARO in colorectal 29 tumorigenesis was evaluated by Park et al. (2001). They created PPARO null cell lines (PPAR5 -/-) by deleting the PPARB gene in a human colon cancer cell line using targeted homologous recombination. When inoculated as xenografis in nude mice, PPARO -/- exhibited a decreased ability to form tumors compared with PPAR5 +/— and wild-type controls, suggesting that suppression of PPARO expression contributes to the growth-inhibitory effects of the APC tumor suppressor (Park et al., 2001) Recent immunohistochemical analysis of B-catenin showed that B-catenin staining was mainly localized to the cell membrane in normal epithelial cells of stomach, duodenum, appendix, and rectum, whereas B-catenin accumulated in the cytoplasm and/or nucleus in 57 cases (79.2 %) out of 72 cases of human gastrointestinal carcinoid tumors (Fujimori et al., 2001). McEntee et al. (1999) reported that sulindac regressed small intestinal adenomas and reduced B-catenin expression in adenomas of ApcMm mice. Mutanen et al. (2000) also reported that rye bran diet prevented tumor formation in ApcMin by decreasing the cytosolic B-catenin level. Sheng et al. (1998) found nuclear localization of B-catenin in hereditary and carcinogen-induced intestinal adenomas. B-catenin was expressed in the cell membrane and cytoplasm of morphologically normal intestinal cells whereas it was found in the nucleus of adenomas from ApcMin mice and Apc 716 mice. Cellular distribution of B-catenin was also modified by sphingolipid consumption (Schmelz et al., 2001). Sphingolipid feeding reduced adenoma numbers in all regions of intestine Min in Apc mice and sphingosine reduced cytosolic and nuclear B-catenin and 30 inhibited growth of human colon cancer cell lines SW 480 and T84 (Schmelz et al, 2001). He et a1 (1999) showed that sulindac sulfide repressed PPARO activity in HCT116 and SW480 cell lines in a dose-dependent manner. A proposed model for APC- and NSAID-mediated suppression of intestinal tumorigenesis (Fig. 5) shows several features of NSAID-mediated chemopreventive action. First, numerous studies have shown that NSAIDs inhibit COX activity and subsequently decrease levels of prostaglandins. However, apoptosis-inducing activities of NSAIDs are not entirely related to the inhibition of COX, suggesting additional cellular targets. Second, NSAIDs inhibit PPARS activity (He et al., 1999). The pro-apoptotic effect of NSAIDs may be explained by direct inhibition of PPARO. Third, NSAID-induced regression of intestinal adenomas is associated with reduced levels of B-catenin (McEntee et al., 1999). McEntee et a1. (1999) observed a 50% M'" mice decrease in B-catenin as well as Bcl-2 in small intestinal tumors from Apc treated with sulindac. Rosin-Arbesfeld et al. (2000) identified a nuclear export function of the APC gene. They showed that APC contains highly conserved nuclear export signals 3’ and adjacent to the mutation cluster region that enable it to export B-catenin from the nucleus. When this ability is lost in APC mutant cancer, the result is accumulation of B-catenin in the nucleus. 31 Sulindac NSAIDs Lipids sPLA2 l '— TCF-4/B-catenin Arachidonic Acid LOX / \ COX l__ ‘A Leukotrienes Prostaglandins PPAR5 CLND] c-MYC MMP 7 Altered Eicosanroid Metaboliirg Figure 5. APC-and NSAID-mediated suppression of intestinal tumorigenesis (Modified from He et al., 1999) 32 Hypotheses and Research Objectives Tart cherry anthocyanins may suppress intestinal tumorigenesis because they inhibit the activities of COX enzymes. However, the potential of the anthocyanins and cyanidin to inhibit colon cancer development has not been fully established. Few studies have examined the chemopreventive potential of anthocyanins and cyanidin. Cyanidin inhibited epidermal growth factor receptor (EGF R) kinase activity in cancer cells (Meiers, 2001). Treatment with an irreversible inhibitor of the EGFR- kinase reduced tumor development in ApcMin mice (Torrance et al., 2000). Hagiwara et al. (2001) demonstrated that anthocyanins in purple corn color reduced the promotion of colon tumors caused by PhIP in rats initiated with dimethylhydrazine. However, they did not test the potential of purple corn color anthocyanins to directly suppress tumor development (Hagiwara et al, 2001). The overall objective of the study is to determine the effects of tart cherry anthocyanins on the development of intestinal adenomas and on global gene Min expression in intestine, cecum and colon of Apc mice. The hypotheses to be tested and specific aims are: l. Tart cherry anthocyanins inhibit intestinal tumori genesis in ApcMin mice. Specific aims: a. Examine ontogeny of intestinal adenoma development in mouse model (Apc mouse) to obtain baseline data on tumor development. b. Examine effects of tart cherry anthocyanins, their aglycone, cyanidin, and their degradation products on growth of human colon cancer cell lines HT 29 and HCT 116. c. Examine effects of tart cherry diets, anthocyanins and their aglycone, cyanidin on multiplicity and size of intestinal adenomas in ApcM'" mice. Min 33 2. The inhibiton of intestinal tumorigenesis caused by tart cherry anthocyanins is M'" mice. related to changes in gene and protein expression in Apc Specific aims: a. Examine the exaression and cellular distribution of [Pi-catenin in intestinal sections of Apc '" mice fed tart cherry anthocyanins in comparison to Apc mice fed control or sulindac-supplemented diets. b. Examine the expression of cancer-related genes in intestinal epithelial cells of ApcMm mice fed tart cherry anthocyanins in comparison to ApcMm mice fed control or sulindac-supplemented diets. I will specifically determine the expression of the predicted target genes of the Wnt/B-catenin signaling pathway such as PPARS, C-Myc, CLND] and MMP7. Min The ApcMin mouse strain is an excellent model for studying colon cancer development and the anticarcinogenic potential of dietary factors. The time frame of tumor development in ApcMin mice will influence experimental protocols. First, we will examine the timing of small intestinal and colonic tumor development in ApcM'n mice and determine the influence of sex on small intestinal and colonic adenoma Min multiplicity and size in Apc mice. We will then determine the effects of tart cherry anthocyanins, cyanidin and sulindac on intestinal adenoma multiplicity and size in the small intestine, cecum and colon in ApcMin mice. The ability of tart cherry anthocyanins to directly inhibit the growth of human colon cancer cell lines HT 29 and HCT 116 also will be determined. Expression of thousands of genes can be examined simultaneously using a novel technique called cDNA microarray analysis. cDNA microarrays are capable of profiling gene expression patterns of tens of thousands of genes in a single experiment (Duggan et al., 1999). Microarrays consist of a collection of immobilized genes which can be simultaneously analyzed to ascertain their expression in tissue. 34 DNA targets, in the form of 3’ expressed sequence tags (ESTs), are arrayed onto glass slides and probed with fluorescent-labeled cDNAs. Premade microarrays of gene-specific cDNA fragments immobilized on glass slides for the mouse will be used to analyze 15,000 genes that survey major biological pathways (Tanaka et al., 2000) Screening changes in expression of such target genes as well as oncogenes and tumor suppressor genes in the different sections of intestine may yield insights about the mechanisms responsible for tumor modulation by anthocyanins and sulindac. We speculate anthocyanins may inhibit expression of PPARB, c-MYC, CLNDl or matrilysin and influence B-catenin expression and distribution in the cell membrane, cytoplasm and nucleus in the intestinal cells. 35 CHAPTER III ONTOGENY OF INTESTINAL ADENOMA DEVELOPMENT IN APCM'N MICE. 36 ABSTRACT ApcMin mice (C57BL/6J Apcmmh) develop small intestinal and colonic adenomas spontaneously. This experiment was conducted to determine the influence of age on the numbers and sizes of small intestinal, cecal and colonic tumors in ApcMin mice. ApcMin mice were weaned at 28 days of age and fed Teklad 8640 rodent diet throughout the experiment. Mice were sacrificed at 30, 45, 60, 75, 90 and 120 days of age. The entire small intestine, cecum, and colon were removed and the small intestine was separated into proximal, middle and distal segments. Intestinal sections were fixed in 10% neutral buffered formalin and stained with 0.2% methylene blue. Intestinal tumors were counted and measured. Body weights and the lengths of the small intestine and the colon increased with age. The numbers and sizes of adenomas in the small intestine also increased with age (p<0.05). The numbers of adenomas in the cecum and colon did not increase significantly with age, but the volume of cecal tumors was not influenced by age. Colonic tumors increased significantly after 75 days of age. Sex did not influence the numbers of small intestinal or colonic tumors. Mouse body weights, colon length, and colon tumor Min volume were influenced by sex. Male Apc mice had greater body weights, colon lengths and colon tumor volumes than female ApcMin mice. Tumors were already present in 30-day-old mice. These results indicate that intestinal tumors develop early in life and progress in size with increasing age in ApcMin mice. Experiments examining the effects of dietary or other interventions in ApcMin mice should take into account their early and rapid tumor development. 37 INTRODUCTION Colon cancer is the second most common cause of cancer mortality and the fourth most common in incidence in the United States (American Cancer Society, 2000). It has been estimated that 66-75% of colon cancer could be preventable by proper diets (WICR/AICR, 1999). Hence, there has been great interest in studying the effects of dietary or other interventions on colon carcinogenesis. Most research on colon cancer prevention by diet has been conducted using animal models of carcinogenesis. Most of these studies rely on tumors initiated by injecting large doses of carcinogens and ofien have used different experimental conditions (i.e. different carcinogens, injection doses, etc). This makes it difficult to compare results from studies conducted in different laboratories. The ApcMin mouse has been proposed to be a model for the study of human colorectal cancer (Moser et al., 1990). ApcMin rrrice are a mutant mouse lineage predisposed to multiple intestinal neoplasia (Min) resulting from a mutation in the murine homolog of the adenomatous polyposis coli (APC) gene (Su et al., 1992). The APC gene also is frequently mutated in humans who develop sporadic colon cancer and is the mutation responsible for causing familial adenomatous polyposis (FAP), an autosomal dominantly inherited disease that predisposes persons to colorectal cancer. The primary phenotype of ApcMin mice is the development of multiple adenomas which progress to adenocarcinomas of the intestine in older mice. Min is transmitted by affected mice to 50% of progeny with an unbiased sex distribution, as is characteristic of a fully penetrant autosomal dominant trait (Moser et al., 1990). 38 The Min mouse strain may be an excellent model for studying colon cancer development and the anticarcinogenic potential of dietary factors. However, the time frame of tumor development in ApcMi" needs to be more thoroughly investigated to allow optimization of experimental protocols. Therefore, the purpose of this study was to examine the timing of small intestinal and colonic tumor development in Min Apc mice and to determine the influence of sex on small intestinal and colonic Min adenoma multiplicity and size in Apc mice. 39 MATERIALS AND METHODS Animals and Diets. All research was conducted with approval of the MSU All University Committee on Animal Use and Care. Mice were housed in MSU University Laboratory Animal Resources maintained facilities. A colony of ApcMin mice was maintained by crossing carrier (C57BL/6J ApcMm/T ) male mice with normal adult female C57BL/6J mice. Mice were housed in a temperature and humidity-controlled room (20-22 °C, 50%) with a 12-h light/dark cycle. Mice were weaned at 28 days of age and fed Teklad 8640 rodent diet throughout the experiment. Mice were not genotyped before being randomly assigned to study groups (ages). We anticipated that 50% of the mice allotted to each group would carry the ApcM'" mutation and 50% would be normal with respect to Apc gene status. Body weights were measured when mice were sacrificed at 30, 45, 60, 75, 90 and 120 days of age. Tumor Number and Size. Upon sacrifice, a section of liver was removed and frozen immediately for Apc genotyping using a PCR-based procedure (Su et al., 1992) followed by gel electrophoresis. The intestine was removed and tissues were separated into the following sections: proximal one-third of small intestine, middle one-third of small intestine, distal one-third of small intestine, cecum, and colon. All intestinal sections were opened longitudinally, rinsed thoroughly with water, fixed in neutral-buffered formalin, and then stained with 0.2% methylene blue. Tumor numbers and sizes were determined in each intestinal segment by direct counting with the aid of a dissecting microscope. Because small intestinal tumors are typically sessile and cecal and colonic tumors are typically polypoid, the tumor sizes were determined by measuring the mean diameter of flat tumors in the small 40 intestine and by measuring volume of three-dimensional tumors in the cecum and colon. Spheric volumes were calculated for cecal and colonic tumors using the formula V = 0.523(width x length x height). Tumor numbers in each small intestinal segment were summed to obtain a total small intestinal tumor burden for each mouse. Body weights, tissue lengths, tumor numbers and tumor diameters in the small intestine were analyzed by two-way analysis of variance to detect the effects and potential interactions of age and sex. For tumor numbers and volumes in the cecum and colon, data were transformed to ranks and then analyzed by two—way analysis of variance (AN OVA). When significant treatment effects were detected (P<0.05), means were compared using the Least Significant Difference (LSD) method. 41 RESULTS Body weights of mice were influenced by age, sex, and Apc status (Min vs. normal). Body weights of all mice increased with increasing age (p<0.05) (Fig. 6a). Male mice weighed approximately 3 grams heavier than female mice for ages 45 through 120 days. Body weights of ApcMin mice (Fig. 6b) averaged about 1 gram less than those of normal Apcm mice at all ages. Small intestine and colon length increased with age (p<0.05) (Table 1). ApcMin mice had small intestine lengths about 9 mm less than normal Apcm mice. Small intestine length was not influenced by sex. However, colons were approximately 4 mm longer in males than females. Carrier status did not influence colon length. Min Intestinal tumors were only detected in Apc mice. Tumor numbers (Fig. 7) in the small intestine of ApcMin mice also increased with age (p<0.05). Mice averaged 13.6 small intestinal tumors at 30 days of age. Small intestinal tumor numbers increased steadily to a maximum of 70-80 tumors per mouse by 90 days of age. As mice aged, the numbers of adenomas in the distal small intestine increased to a greater magnitude compared to those in the proximal and middle small intestine. The proportion of small intestinal adenomas in proximal, middle, and distal segments was 28, 34, and 38%, respectively, in 30-d old mice and 10, 34, and 56%, respectively, in 120-d old mice. The average diameter of the small intestinal adenomas (Fig. 8) was relatively constant from 30-75 days of age, averaging 0.94 mm per tumor. This diameter increased significantly in 90-d old mice (1.2 mm) and increased dramatically in 120—d old mice to 1.8 mm per tumor. There was no sex effect on small intestinal tumor numbers or diameters. 42 a) -o—All females b) —0— Min females +All males -+—Min males 30 30 25 25 m 20 20 (I) E 15 E 15 -- o (’5 10 - 10 . 5 l- 5 .. 0 1 1 1 1 0 1 4 I r 30 45 60 75 90120 30 45 60 75 90 120 Age (Days) Age (Days) Figure 6. Influence of age on body weights a) Influence of age on body weights of all mice. Each point represents the mean of the age groups (30 days; n=22, 45 days; n=l7; 60 days; n=l7; 75 days; n=22; 90 days, n=23,l20 days; n=23). Body weight increased with age at all time points except between 60 and 75 days. Error bars indicate SEM. Males were significantly larger (p<0.05) than females after 30 days of age. b) Influence of age on body weights of ApcMin mice. Each point represents the mean of the age groups (30 days; n=8, 45 days; n=10; 60 days; n=10; 75 days; n=9; 90 days, n=9,120 days; n=8). Error bars indicate SEM. Body weights increased with age at all time points except between 60 and 75 days. Body weights were significantly different between males and females after 30 days of age. 43 Table 1. Lengths of small intestine and colon of all mice (30 days; n=22, 45 days; n=l7, 60 days; n=l7, 75 days; n=22, 90 days; n=23,120 days; n=23) (Normal; n=54, Min, Apc , n=70) (Female; n=56, male; n=68). Each value represents mean i SEM. Means in the same column within a category not sharing a common superscript are significantly different (p<0.05). Age (days) Small intestine (mm) Colon (mm) 30 308 i 42‘ 68 i 2 a 45 319 i 5 a 76 :1: 2 b 60 339:4b 79:2bc 75 349i4b 80:2bc 90 349 a: 4 b 78 i 2 b 120 378:4c 84:2c Apc Status Small intestine (mm) Colon (mm) Normal 345 i 2 b 78 i 1 Min 336 i 2 a 78 i 1 Sex Small intestine (mm) Colon (mm) Female 338 i 2 75 1r 13 Male 343 i 2 80 i 1 b 44 LC] Proximal I Middle Distal I Total i 100 90 T” d cd 80 c 70 - 60 ~ 50 ~~ o Number 4o 4 7 b/ 30 . g 20 - {13% 1o -- 1? 0 _ I1? 90 120 Figure 7. Influence of age on adenoma numbers in proximal (white), middle (gray) and distal (hatched) segments of the small intestine and total (black) numbers in the small intestine (30 days; n=8, 45 days; n=10; 60 days; n=10; 75 days; n=9; 90 days, n=9,120 days; n=8). Error bars indicate SEM. Columns of the same shade not sharing a common superscript are significantly different (p<0.05). 45 1.8 *- 1.6 4 1.4 a 1.2 ‘— T Millimeters 0.8 *- 0.6 4 0.4 ~ 0.2 ~— I 30 45 . 60 75 90 120 Age (Days) Figure 8. Influence of age on the average diameter of small intestinal adenomas (30 days; n=8, 45 days; n=10; 60 days; n=10; 75 days; n=9; 90 days, n=9, 120 days; n=8). Error bars indicate SEM. Columns not sharing a common superscript are significantly different (p<0.05). 46 The numbers (Fig. 9) of adenomas in the cecum were not significantly influenced by age. The mean volume of cecal adenomas was larger in older mice (Fig. 10), but this effect did not reach statistical significance. There was no sex effect on cecal tumor number or volume. The number of adenomas in the colon (Fi g. 9) was influenced by age, but not in a consistent manner. Mice of 90-d of age had significantly more colonic tumors than mice at 45 or 60 days. The total volume of colonic tumors was significantly influenced by age (Fig. 11). Colonic tumors were small in size through 75 days of age, and then increased in volume dramatically after this time. There was also a significant sex effect on colon tumor volume. Colon adenoma volume averaged 28.9 mm3 larger in males (43.8 mm3) than in females Min (14.9 mm3) at 120 days of age. This sex effect was not detected in the Apc mice from the subsequent studies. 47 ll Cecum I Colon : Number 3O 45 60 75 90 120 Age (Days) Figure 9. Influence of age on the average numbers in the cecum (gray) and colon (black) (30 days; n=8, 45 days; n=10; 60 days; n=10; 75 days; n=9; 90 days, n=9, 120 days; n=8). Error bars indicate SEM. Columns of the same shade not sharing a common superscript are significantly different (p<0.05). 48 2.5 _- 0.5 —~ Cubic Millimeters/mouse 30 45 60 75 90 120 Age (Days) Figure 10. Influence of age on adenoma volume (mm3) in the cecum (30 days; n=8, 45 days; n=10; 60 days; n=10; 75 days; n=9; 90 days, n=9, 120 days; n=8). Error bars indicate SEM. 49 Cubic Millimeters/mouse 30 45 60 75 90 1 20 Age (Days) Figure 11. Influence of age on adenoma volume (mm3) in the colon (30 days; n=8, 45 days; n=10; 60 days; n=10; 75 days; n=9; 90 days, n=9, 120 days; n=8). Error bars indicate SEM. Columns not sharing a common superscript are significantly different (p<0.05). 50 DISCUSSION Mouse models such as the Min mutant should provide a major resource for studying the anticarcinogenic potential of dietary factors, pathways of tumorigenesis, and the molecular identification of genes that can control particular neoplastic processes. The Min mouse has been and will continue to be used as a model to study M' - '" mice effects of dietary factors on human colorectal cancer. Effective use of Apc in such studies requires a thorough assessment of their normal process of tumor development. Min A few concerns have been identified concerning the use of Apc mice as a model for research of colorectal cancer. Unlike human intestinal cancers, which are Min predominantly colorectal, Apc mice develop the majority of their tumors in the small intestine. The utility of ApcMin mice may also be limited by their lack of large numbers of colonic adenomas and aberrant crypt foci, the putative precursors of large-bowel polyps and cancers. These potential limitations also would pertain to various other Apc knockout mouse strains such as Apc‘m, ApcA716 and Apc474 (Fodde et al., 1994; Oshima et al., 1995; Sasai et al., 2000). Accelerated polyposis models have been deveIOped to increase the observed numbers of tumors and aberrant crypts. For example, additional carcinogen injections (Shoemaker et al., 1995; Steffensen et al., 1997) or genetic mutation in mismatch repair genes have been added to ApcMin (Baker et al., 1998; Lal et al., 2001). However, the majority Min still uses the standard Apc mouse model for the study of colorectal cancer development. 51 Realtively little research has been conducted to determine the tumor development characteristics of ApcMin mice. Shoemaker et al. (1995) have shown age-related effects on the formation of adenomas in ApcMin mice after additional treatment with ethylnitrosourea (ENU). Their results demonstrate that ENU-induced Min intestinal tumors in Apc mice are most likely to be initiated during the first 2 weeks of life. In the present study, the time frame of tumor development in Min mice was determined. Our results provide baseline data for the influence of age and sex on Min body weight and intestinal tissue length of Apc mice in comparison with those of normal Ape”+ mice. These results also describe the influence of age and sex on adenoma development of ApcMin mice. Small intestinal, cecal and colonic adenomas were already present in 30 day-old mice. Tumor volumes remained relatively small throughout the intestine for the first 75 days of age. After this time, tumor volumes increased rapidly up to 120 days of age. We chose 120 days as the final time point in this study because it has been observed previously that ApcMin mice suffer considerable morbidity and mortality beyond this age (Moser, 1990). Morbidity and mortality in ApcMin mice is typically due to anemia caused by hemorrhraging associated with the small intestinal tumors after they progress beyond a certain size. We have observed that there is little Mi“ mice that have small intestinal tumors less than 1.5 mm in morbidity in Apc diameter. However, small intestinal tumors greater than 1.5-2.0 mm in diameter tend to perfolate and hemorrhrage. Hence, the choice of an end point (age) for feeding studies conducted using ApcMin mice must be fully considered. Tumor progression 52 occurs predominantly in the age range of 75-120 days, with significant morbidity observed thereafter. These results suggest that experiments examining the effects of dietary or other interventions in ApcMin mice should take into account their early and rapid tumor development. 53 CHAPTER IV TART CHERRY ANTHOCYANINs INHIBIT CECAL TUMOR DEVELOPMENT IN APCMIN MICE AND REDUCE GROWTH OF HUMAN COLON CANCER CELLS 54 ABSTRACT Anthocyanins, which are bioactive phytochemicals, are widely distributed in plants and especially enriched in tart cherries. Based on previous observations that tart cherry anthocyanins and their respective aglycone, cyanidin, can inhibit cyclooxygenase enzymes, we conducted experiments to test the potential of anthocyanins to inhibit intestinal tumor development in ApcMin mice and growth of human colon cancer cell lines. Mice consuming the cherry diet, anthocyanins, or cyanidin had significantly fewer and smaller cecal adenomas than mice consuming the control diet or sulindac. Colonic tumor numbers and volume were not significantly influenced by treatment. Anthocyanins and cyanidin also reduced cell grth of human colon cancer cell lines HT 29 and HCT 116. The IC50 of anthocyanins and cyanidin was 780 11M and 63 uM for HT 29 cells, respectively and 285 1.1M and 85 uM for HCT 116 cells, respectively. These results suggest that tart cherry anthocyanins and cyanidin may reduce the risk of colon cancer. 55 INTRODUCTION Tart cherries contain substantial quantities of anthocyanins in addition to other bioflavonoids (Wang et al., 1997). Anthocyanins, a member of the bioactive phytochemicals, are widely distributed in fi'uits, vegetables and beans, suggesting that plant-based diets can provide considerable amounts of anthocyanins (Wang et al., 1999; Seeram et al., 2001). Like the vast majority of flavonoids, anthocyanins primarily occur in plants as glycosides. Cyanidin is the major anthocyanin aglycone in tart cherries. Montmorency and Balaton;rM tart cherries contain 0.40 to 0.80 mg/ g, respectively, of anthocyanins (Wang et al., 1997). These anthocyanins were found to function as antioxidants and cyanidin was shown to inhibit the activities of COX enzymes in vitro (Wang et al., 1999a; Wang et al., 1999b). Several studies have demonstrated that nonsteroidal anti-inflammatory drugs (N SAIDs) inhibit the growth of colon tumors in animal models and reduce the risk of colon cancer in humans (Giovannucci et al., 1995; Labayle et al., 1991). In most cases, colon carcinogenesis depends on mutation of the APC gene, which is considered a gatekeeper in the carcinogenic process (Kinzler and Vogelstein, 1996). Human APC gene germline mutations cause familial adenomatous polyposis, an autosomal dominantly inherited disease that predisposes affected individuals to develop numerous adenomatous polyps and, ultimately, colorectal cancer (Su et al., 1992). APC gene mutations also are a frequent and early event in sporadic colon cancer. ApcMin mice are a mutant mouse lineage predisposed to multiple intestinal neoplasia (Min) due to a mutation in the murine homolog of the adenomatous polyposis coli (APC) gene (Moser et al., 1990). The primary phenotype of ApcMin mice is the 56 development of multiple intestinal adenomas that progress to adenocarcinomas of the intestine in older mice. The objectives of this research were to determine the potential of tart cherry anthocyanins and cyanidin to inhibit intestinal tumor development in ApcMin mice and to determine the potential of anthocyanins and cyanidin to directly inhibit the grth of human colon cancer cells. 57 MATERIALS AND METHODS Animals and Diets. This research was conducted with approval of the Michigan State University All-University Committee on Animal Use and Care. Mice were housed in a temperature and humidity-controlled room (20-22 °C, 50%) with a 12-h light/dark cycle. ApcMin progeny were identified by a PCR-based assay (Su et al, 1992) and were randomly assigned to five treatment groups (n = 10 per group; equal numbers of males and females) at 4 - 5 weeks of age and fed treatment diets for 10 weeks. Deionized water and diets was provided ad libitum. The treatments (Table 2) were; 1) control diet (modified American Institute of Nutrition 93G diet containing 220 g/kg protein, 150 g/kg soybean oil and 50 g/kg cellulose), 2) control diet + 800 mg/L anthocyanins in the drinking water, 3) control diet + 200 mg/L cyanidin in the drinking water, 4) control diet + 200 mg/L sulindac (an NSAID) in the drinking water, and 5) modified control diet containing 200 gfkg freeze—dried pitted tart cherries. Cherries were frozen, freeze-dried and ground before they were incorporated into the diet. Anthocyanins were isolated from tart cherries and were a mixture of 3-cyanidin 2"-0-,B-D-glucopyranosyl-6"-0-a-L- rhamnopyransyl-fl-D-glucopyranoside and 3-cyanidin 6"-0-a—L-rhamnopyranosyl- ,B—D-glucopyranoside (Wang et al, 1999) at 65 and 35%, respectively. The aglycone, cyanidin, was prepared from the anthocyanins (Wang et al, 1997). Ascorbic acid (50 mg/L) was added to the drinking water of all mice to enhance the stability of anthocyanins and cyanidin in solution by lowering the pH. 58 Table 2. Composition of the experimental diets (Percentage of diet) Ingredient Modified AIN-93G diet 20% Cherries diet Casein 22.12 22.12 Soybean Oil 15.00 15.00 Corn Starch 31.72 24.22 Dyetrose 10.57 8.07 Sucrose 10.00 0.00 AIN-93G-MX 3.87 3.87 AIN-93G-VX 1.11 1.11 L-Cystine 0.33 0.33 Choline Bitartrate 0.28 0.28 Tert-Butylhydroquinone 0.003 0.003 Cellulose 5.00 5.00 Dried Cherries 0.00 20.00 59 Tumor Number and Size. The mice were sacrificed after 10 weeks of treatment and the numbers and sizes of adenomas in the intestinal sections were measured. The entire small intestine, cecum, and colon were removed from each mouse to determine the number and size of adenomas. Intestinal sections were opened longitudinally, rinsed thoroughly with water, fixed overnight in 10% neutral-buffered formalin, and then stained with 0.2% methylene blue. Tumor numbers and dimensions for each intestinal segment were determined by direct counting with the aid of a dissecting microscope and measuring grid. The tumor sizes were determined by measuring the spherical (three dimensional) volume of adenomas in the cecum and colon and the average diameter of tumors in the small intestine. Tumors in the cecum and colon of ApcMin mice typically are polypoid in appearance, whereas the small intestinal tumors are sessile. Spherical volumes of cecal and colonic tumors were calculated by the formula: Volume = 0.532 x (width x length x height of tumor). Tumor numbers and diameters in the small intestine were analyzed by two- way analysis of variance (treatment, sex). For tumor numbers and volumes in cecum and colon, data were transformed to ranks and then ranks were analyzed by two-way analysis of variance. When significant treatment effects were detected (P<0.05), treatment means were compared using the Least Significant Difference method. Six mice (one from the control group, two from the anthocyanin group, and three from the cyanidin group) were excluded from the final statistical analysis because it was determined at the end of the experiment that these animals did not carry the APCMin gene mutation, and therefore did not develop intestinal tumors. 60 Cell Culture and Growth Assays. The human colorectal cancer cell lines HCT 116 and HT 29 (American Type Culture Collection) were cultivated in McCoy’s 5A media supplemented with 10% fetal bovine serum. Cells were harvested for growth assays when they had reached 50-80% confluence by trypsianDTA treatment and counted using a hemacytometer. Cells were then seeded at 15 ,000 cells/well in 24-well tissue culture plates. Plates were incubated overnight at 37°C and 5% CO; to allow cells to attach and begin proliferating. At the beginning of treatments, the media was gently aspirated from each of the wells, which were then rinsed with PBS. One ml of treatment media was added to each well (n = 8-12 wells per treatment level per cell line) and plates were incubated for 72 h. Treatment media was McCoy’s 5A media supplemented with 10% fetal bovine serum and containing the respective concentrations of anthocyanins (0-1000 11M) or cyanidin (0-250 uM). Anthocyanins were dissolved in distilled water before addition to the treatment media, whereas cyanidin was solubilized in DMSO before addition. When used, the DMSO concentration was equalized for all treatment media and never exceeded 0.1% (v/v) of the final treatment media. Total cell numbers in each well were quantified after 72 h of incubation in treatment media. Cell numbers were calculated based on total DNA content in each well using a procedure that quantifies DNA based on fluorescence of bound Hoescht 33258 (Labarca et a1, 1980). Fluorescence was measured by a Cytofluor II fluorimeter (Applied Biosystems; Foster City, CA) using an excitation wavelength of 360 nm and an emission wavelength of 460 nm. Fluorescence readings were converted to DNA by comparison to standard solutions of Salmon testis DNA 61 (Sigma Chemical Company; St. Louis, MO). The cell numbers in each well were calculated by converting the quantity of DNA in each well by the amount of DNA present in each cell (determined experimentally) for HCT 116 and HT 29 cells. Cell numbers observed in each well after 72 h of growth were corrected for initial cell number (determined at the time treatment media was added). These data were then subjected to multiple regression analysis to develop least-squares polynomial equations describing the influence of anthocyanins or cyanidin concentration on cell number. These equations were then used to iteratively calculate the concentration of anthocyanins or cyanidin required to cause a 50% reduction (ICso) in grth (cell number) for each cell line. 62 RESULTS Anthocyanins, Cyanidin and Tart Cherries Influence Intestinal Tumor Development. Final body weights of mice were significantly influenced by treatment and averaged 22.8, 24.1, 21.3, 19.7, and 25.5 grams for mice consuming control diet, anthocyanins, cyanidin, tart cherries, and sulindac, respectively (Fig. 12). Final body weights for mice consuming anthocyanins and sulindac were greater (P<0.05) than for mice consuming tart cherries. Averaged across all treatments, male mice weighed significantly greater (P<0.05) than female mice both at the beginning (18.3 versus 15.6 grams) and the end (24.6 versus 20.8 grams) of the experiment. Male mice weighed approximately 4 grams heavier than female mice. Treatments had differential effects on tumor incidence and burden in the various sections of the intestinal tract. Mice consuming anthocyanins, cyanidin, or tart cherries had fewer (P<0.05) adenomas in the cecum than mice consuming the control diet or sulindac (F ig. 13). The average number of adenomas in the cecum of mice that consumed anthocyanins, cyanidin and tart cherries was 0.6, 0.6, and 0.6, respectively, whereas mice that consumed the control diet or sulindac had an average of 2.4 and 3.8 cecal tumors, respectively. Colonic adenoma number was not influenced by treatment (Fig. 13). Average adenoma numbers in the colon were 3.6, 3.1, 3.6, 3.3, and 5.2 for mice that consumed the control diet, anthocyanins, cyanidin, tart cherries, and sulindac, respectively. The total burden (volume) of cecal adenomas was less (P<0.05) in mice consuming anthocyanins, cyanidin or tart cherries when compared to mice consuming the control diet or sulindac (Fig. 14). Adenoma volumes in the cecum of mice that consumed anthocyanins, cyanidin, or 63 tart cherries were 0.7, 0.6, and 1.8 mm3 respectively, whereas those in mice consumed the control diet and sulindac were 3.0 and 4.0 mm3, respectively (Fig. 13). Although mice that consumed tart cherries had the greatest adenoma burden in the colon (8.4 m3), this was not statistically greater than that observed in mice consuming the other treatments (Fig. 14). Tumor multiplicity in the small intestine was not significantly influenced by treatment and averaged 48 tumors per mouse (Fig. 15). Mice that consumed sulindac ' had the smallest number of small intestinal adenomas (28 per mouse), but this was not statistically different than small intestinal tumor numbers observed for the other treatments. The average size of small intestinal adenomas (Fig. 16) was increased (P<0.05) by feeding tart cherries (1.7 mm) and reduced (P<0.05) by sulindac (0.9 mm) relative to that observed in mice consuming the control diet (1.4 mm), anthocyanins (1.3 mm), or cyanidin (1.3 mm). l l _._Contr0| —o—Anthocyanins ' ‘ " ' Cyanidin ii i- + — Sulindac —II— Cherry 30 Figure 12. Influence of anthocyanins, cyanidin, tart cherries and sulindac on body weights (g) of mice throughout the experimental period. Error bars indicate SEM. 65 {I Cecum I Colonj Adenoma Number E33 Anthocyanins Cyanidin Cherries Sulindac Figure 13. Influence of anthocyanins, cyanidin, tart cherries and sulindac on adenoma numbers in cecum (gray) and colon (black). Error bars indicate SEM. Columns of the same shade not sharing a common superscript are significantly different (P<0.05). 66 1E Cecum I Colon ] 12.0 10.0 ~— 8.0 + Cubic millimeters . 2.241 at Control Anthocyanins Cyanidin Cherries Sulindac Figure 14. Influence of anthocyanins, cyanidin, tart cherries and sulindac on adenoma volumes (mm3) in cecum (gray) and cOlon (black). Error bars indicate SEM. Columns of the same shade not sharing a common superscript are significantly different (P<0.05). 67 too 90 - 80 - 70 ~ so 4 50 . 4o - 30 - 20 - 1o 4 O - Adenoma Number Control Anthocyanins Cyanidin Cherries Sulindac Figure 15. Influence of anthocyanins, cyanidin, tart cherries and sulindac on adenoma numbers in small intestine. Error bars indicate SEM. 68 -L m N 1 T 1.61 fl I T Adenoma Diameter (mm) .0 .O .0 _o 7" I" O N -§ 0) m —* N A 1 1 1 % 1 H 1 1 Control Anthocyanins Cyanidin Cherries Sulindac Figure 16. Influence of anthocyanins, cyanidin, tart cherries and sulindac on average diameters (mm) of adenomas in small intestine. Error bars indicate SEM. Columns not sharing a common superscript are significantly different (P<0.05). 69 Anthocyanins and Cyanidin Inhibit Human Colon Cancer Cell Growth. Treatment with anthocyanins (Fig. 17) or cyanidin (Fig. 18) caused a dose-dependent reduction in cell numbers for both HCT 116 and HT 29 cells. Neither anthocyanins nor cyanidin caused cytotoxicity even at the highest concentrations tested, as indicated by little or no dead cells. Cyanidin was far more effective in inhibiting the grth of these cancer cell lines than anthocyanins. The observed IC50 values for cyanidin were 85 and 63 1.1M for HCT 116 and HT 29 cells, respectively, whereas those for anthocyanins were 260 and 585 11M for HCT 116 and HT 29 cells, respectively. 70 EJHCT116 IHT29' 120 100 80 , 60 I 4o . 20 ~ Cell Number (% of control) 0 31.3 62.5 125 250 500 1000 Anthocyanin Concentration (um) Figure 17. Influence of Enthocyanins on growth of human cancer cells, HCT 116 (gray) and HT 29 (black). Error bars indicate SEM. 71 cctccc Co .ch cchzz :60 F rgurt and H' IHCT116 IHT29 140 120 Cell Number (% of control) 20 100 - 60 40 ~ ‘ 0 7.8 15.6 31.3 62.5 125 250 Cyanidin Concentration (um) Figure 18. Influence of cyanidin on growth of human cancer cells, HCT 116 (gray) and HT 29 (black). Error bars indicate SEM. 72 DISCUSSION Our interest in testing the potential of tart cherry anthocyanins and cyanidin Mi“ mice stemmed from the observation that to inhibit tumor development in Apc these compounds inhibit the activities of COX enzymes (Wang et al., 1999). Other studies have demonstrated that sulindac (and other NSAIDs) reduce small intestinal tumor multiplicity and size in ApcMin mice (Boolbol et al., 1996; Chiu et al., 1997; Mahmoud et al., 2000). In this study, we found that anthocyanins, cyanidin, and tart cherries (presumably as a source of anthocyanins) all significantly reduced the number and burden of tumors in the cecum of ApcMin mice. Conversely, sulindac did not influence tumor development in the cecum. None of the treatments tested influenced the numbers of tumors in the small intestine or the numbers or burden of tumors in the colon. Our inability to detect a significant reduction in small intestinal adenoma number by sulindac was likely due to a number of factors, including the relatively small numbers of mice per treatment group and large variations among individual mice in adenoma development. We also have observed that the ability of sulindac to inhibit small intestinal tumor development decreases as ApcMin mice age (Bourquin, L. D., unpublished research). The lack of effect of anthocyanins or cyanidin on colonic tumor development may be a consequence of their metabolism by intestinal bacteria. The average size of small intestinal tumors was smaller in mice consuming sulindac and larger in mice consuming tart cherries when compared to that in mice consuming the control diet, anthocyanins or cyanidin. Progression in the size of Min small intestinal tumors is highly correlated with morbidity in Apc mice. We have 73 observed that significant morbidity and weight loss occur when small intestinal tumors reach an average size of 1.5 mm diameter. At this stage, the tumors tend to hemorrhage and may perforate the small intestine. The observed differences in final body weight in this experiment likely are a consequence of these differences in small intestinal tumor promotion. It is well documented that sulindac and other NSAIDs consistently reduce the size of small intestinal tumors in ApcMin mice (Boolbol et al., 1996; Chiu et al., 1997; Mahmoud et al., 2000). The mechanism for the larger small intestinal tumor size in mice consuming rat cherries is not known. The effects of the treatments on tumor development were not consistent throughout the intestinal tract. Sulindac and other NSAIDs typically reduce small intestinal tumor development in ApcMin mice, but often have little effect on tumor development in the cecum and colon (Barnes et al., 1998; also Bourquin, L. D., unpublished research). Mice consuming anthocyanins, cyanidin and tart cherries had fewer and smaller tumors in the cecum, but these compounds did not afford any protection to the small intestine. These results indicate that although anthocyanins and cyanidin inhibit the activities of COX enzymes in vitro (Wang et al., 1999; Seeram et al., 2001), they likely do not influence intestinal tumor development in ApcMin mice through a pathway involving COX inhibition. Both anthocyanins and cyanidin inhibited the growth of the colon cancer cell lines HT 29 and HCT 116, although cyanidin was much more effective. We have identified three degradation products from anthocyanins and cyanidin in cell culture medium. These were protocatechuic acid, 2,4-dihydroxybenzoic acid, and 2,4,6- trihydroxybenzoic acids (Seeram et al., 2001). In addition, we have detected trace 74 d1 (11 CC bu qu for sin V11: C01 C01 1'65 P36 quantities of cyanidin—3-glucoside and cyanidin in culture medium after 72 h of cell grth in anthocyanin treatments. We have evaluated the potential of these degradation products to inhibit the growth of HCT 116 and HT 29 cells. None of the degradation compounds assayed demonstrated any inhibition of cell growth at concentrations ranging up to 250 M (Seeram et al., 2001). Anthocyanins and cyanidin are unstable at pH 7.0 and spontaneously degrade to chalcone and benzoic acid derivatives. The red cyanidin cation is stable at pH < 3, but deprotonates and produces ketoquinonoidal bases and finally an ionized quinonoid base at pH > 7 (Seeram et al., 2001). At pH 3—6, the cyanidin cation forms a carbinol pseudobase or chalcone pseudobase. Anthocyanins are highly water-soluble and considered to be structurally similar to a number of strong DNA intercalators (Mas et al., 2000). Both DNA and RNA act as strong copigments for anthocyanins (Mistry et al., 1997). Also, anthocyanins protect DNA against oxidative damage (Sarrna et al., 1999). Under in vivo and cell culture conditions, both anthocyanins and cyanidin potentially form corresponding pseudobases due to pH variations. These pseudobases are transition compounds and may be stable in vivo as protein bound complexes. Given these results, we predict that the anthocyanins, the aglycone cyanidin, or its varying pseudobases directly suppress cell growth and subsequent tumor development. We believe that our results are the first to demonstrate that anthocyanins and cyanidin have the potential to directly interfere with intestinal tumor development. Hagiwara et al. (Hagiwara et al., 2001) demonstrated that anthocyanins in purple corn color reduced the promotion of colon tumors caused by 2-amino-1-methyl-6- 75 phenylimidazo[4,5-b]pyridine (PhIP) in rats initiated with dimethyl hydrazine. However, they did not test the potential of purple corn color anthocyanins to directly suppress tumor development (Hagiwara et al., 2001). In summary, we have demonstrated that tart cherry anthocyanins and their aglycone cyanidin significantly reduced tumor development in the cecum of ApcMin mice. These compounds also directly inhibited the growth of human colon cancer cells in vitro, with the aglycone cyanidin being far more effective than the anthocyanin glycosides. Benzoic acid derivatives yielded from the degradation of anthocyanins and cyanidin had no influence on colon cancer cell growth. Taken together, these results suggest that cyanidin or its corresponding pseudobase is directly inhibiting tumor development in the cecum of ApcMin mice. Anthocyanins also are effective presumably due to their deglycosylation to cyanidin by cecal bacteria. The lack of a clear suppression of tumor development in the colon probably is due to further degradation of the cyanidin molecule by elevated pH in the intestinal lumen and bacterial metabolism. 76 CHAPTER V TART CHERRY ANTHOCYANINS REDUCE INTESTINAL , TUMORIGENESIS AND ALTER INTESTINAL EPITHELIAL CELL GENE EXPRESSION, BUT Do NOT INFLUENCE INTRACELLULAR DISTRIBUTION OF B-CATENIN IN INTESTINE OF APCMIN MICE. 77 ABSTRACT Anthocyanins are flavonoid compounds that impart color to fruits and vegetables including cherries. Previous research in our laboratory has demonstrated the potential of tart cherry anthocyanins to inhibit tumor development in ApcMin mice and reduce proliferation of human colon cancer cell lines. The objective of this research was to determine the extent to which dietary anthocyanins inhibit intestinal tumorigenesis, alter B-catenin expression and distribution, and influence global gene Min expression profiles in intestinal epithelial cells of Apc mice. Mice were randomly assigned to one of three treatment groups at 4 weeks of age (11 = 25 normal + 25 ApcMin mice per treatment). The treatments were 1) AIN 93G control diet, 2) the control diet + 200 mg/kg sulindac, and 3) the control diet + 750 mg/kg anthocyanins. The number of adenomas in the small intestine was reduced (P<0.05) by diets containing either sulindac (:17 = 9) or anthocyanins (X = 17 ) relative to the controls (X = 27). The number and volume of cecal adenomas were reduced (P<0.05) in mice that consumed anthocyanins (0.6, 0.7 m3) compared to mice consuming sulindac (2.1, 4.7 m3) or the control diet (1.0, 1.5 mm3). Mice consuming anthocyanins had significantly fewer colonic adenomas (0.8) than those consuming sulindac (1.6) or the control diet (2.2). Colonic adenoma burden was not influnced by treatment. Dietary anthocyanins reduced tumor development throughout the Min intestinal tract of Apc mice, whereas tumor inhibition by sulindac was specific to the small intestine. Anthocyanins increased (P<0.05) B-catenin mRNA expression in middle small intestine and cecum. Intracellular distribution of B-catenin mRN A was primarily in the epithelial cell membrane for mice on all dietary treatments. cDNA 78 microarray analysis showed that expression of mRNA of B-catenin was not altered by anthocyanins or sulindac. Anthocyanins reduced the expression of cyclin D1 mRNA cOmpared to sulindac in cecum, but expression of mRNA for other predicted target genes of the Wnt/B-catenin signaling pathway - proxisome proliferator activated receptor 6, C-Myc and matrix metalloproteinase 7 — were not altered by anthocyanins or sulindac. These observations suggest that the tumor inhibition caused by feeding anthocyanins or sulindac was not a consequence of any reduction in the cytoplasmic level and translocation of B-catenin to the nucleus and subsequent influences on target gene expression. cDNA microarray analysis identified a total of 1,492 genes that were influenced by any treatment in the small intestine, colon or ceum. Genes influenced by anthocyanins and sulindac are involved in a wide variety of cellular and molecular processes including DNA synthesis and repair, transcription, mRNA processing, translation, protein interactions and turnover, cell proliferation and differentiation, cell cycle control, and programmed cell death. The patterns of gene expression changes suggest that alterations of cellular and molecular M'" mice. processes by anthocyanins and sulindac likely are different in Apc Additional confirmatory analysis of these gene expression results will be necessary to further establish potential mechanisms whereby anthocyanins and sulindac influence tumor development. 79 INTRODUCTION Tart cherries contain substantial quantities of anthocyanins in addition to other bioflavonoids (Wang et al., 1997). Anthocyanins, a member of the bioactive phytochemicals, are widely distributed in fruits, vegetables and beans, suggesting that plant-based diets can provide considerable amounts of anthocyanins (Wang et al., 1999; Seeram et al., 2001). Like the vast majority of flavonoids, anthocyanins primarily occur in plants as glycosides. Cyanidin is the major anthocyanin aglycone in tart cherries. Montmorency and BalatonTM tart cherries contain 0.40 to 0.80 mg/ g, respectively, of anthocyanins (Wang et al., 1997). These anthocyanins were found to function as antioxidants and cyanidin was shown to inhibit the activities of COX enzymes in vitro (Wang et al., 1999a; Wang eta1., 1999b). Several studies have demonstrated that NSAIDs inhibit the growth of colon tumors in animal models and reduce the risk of colon cancer in humans (Giovannucci et al., 1995; Labayle et al., 1991). In previous research, we demonstrated that anthocyanins and cyanidin, when administered in drinking water, reduced the numbers and total burden of tumors in the cecum of ApcMin mice (Chapter IV). Cyanidin and anthocyanins also inhibited the growth of human colon cancer cell lines HT 29 and HCT 116 in vitro (Chapter IV). Min Apc mice are a mutant mouse lineage predisposed to multiple intestinal neoplasia (Min) due to a mutation in the murine homolo g of the adenomatous M' - '" mrce polyposis coli (APC) gene (Su et al., 1992). The primary phenotype of Apc is the development of multiple intestinal adenomas that progress to adenocarcinomas of the intestine in older mice. In most cases, colon carcinogenesis depends on 80 mutation of APC gene and this gene is considered a gatekeeper in the carcinogenic process (Kinzler and Vogelstein, 1996). Most mutations in the human APC gene are found to occur between codons 1286 and 1513, which is within a region encoding B- catenin binding sites (Nakamura, 1993). These mutations result in the production of a truncated APC protein product which cannot facilitate the degradation of B-catenin. Recently, a nuclear export function of the APC gene has been identified (Rosin- Arbesfeld et al., 2000). APC contains highly conserved nuclear export signals 3’ adjacent to the mutation cluster region that enable it to bind and export [3-catenin from the nucleus. When this ability is lost due to APC mutation, B-catenin accumulates in the nucleus. B-catenin functions as a component of the E-cadherin adhesion complex and also as a transcriptional regulator in a signaling pathway by binding to T cell factor (ch) transcription factor in the nucleus. Aberrant accumulation and nuclear localization of B-catenin can enhance trancriptional activation and expression of target genes in this signaling pathway. A few target genes in this pathway — peroxisome proliferator activated receptor 8 (PPARS), c-MYC, cyclin D 1 (CLNDI) and matrilysin (matrix metalloproteinase 7) — have been recently identified (He et al., 1999; He at al., 1998; Tetsu and McCormick, 1999; Crawford et al., 1999). Screening changes in expression of these target genes as well as full genome in the different sections of intestine should yield insights into the potential mechanisms responsible for tumor modulation by anthocyanins and sulindac. 81 Expression of thousands of genes can be examined simultaneously using a novel technique called cDNA microarray. Microarrays consist of a collection of gene-specific cDNA fragments immobilized on glass slides which can be simultaneously analyzed to ascertain their expression in tissue. Anthocyanins and sulindac likely influence tumorigenesis in the intestinal tract by modulating the expression of genes involved in carcinogenesis. The objectives of this study were to determine the extent to which dietary anthocyanins and sulindac 1) inhibit intestinal tumorigenesis, 2) influence the expression and celluar localization of B-catenin, and Min 3) influence global gene expression profiles in intestine of Apc mice. 82 MATERIALS AND METHODS Animals and Diets. This research was conducted with approval of the Michigan State University All-University Committee on Animal Use and Care. Min +/+ Apc mice were produced by mating normal C57BL/6J (Apc ) female mice with Min C5 7BL/6J (ApcMiM) male mice. One-hundred-fifty mice were grouped by sex, weight and litter and then randomly assigned to three treatment groups (n= 50 per group) at 4 weeks of age. Mice were not genotyped to determine Apc status before the experiment, but it was expected that approximately 25 Min mice and 25 normal mice would be randomly allotted to each treatment. Deionized water and diets were provided ad libitum. Body weights were measured weekly. The dietary treatments were; 1) control diet (modified American Institute of Nutrition 93G diet containing 220 g/kg protein, 150 g/kg soybean oil and 50 g/kg cellulose), 2) control diet + 200 ppm sulindac, and 3) control diet + 750 ppm anthocyanins. Sulindac and anthocyanins were incorporated into the appropriate diets upon formulation. The concentrations of sulindac (200 ppm) and anthocyanins (750 ppm) were based on efficacious concentrations used in a previous experiment conducted in our laboratory (Chapter IV). Anthocyanins were isolated from tart cherries and were a mixture of 3-cyanidin 2"-0—,B-D-glucopyranosyl-6"-0-a—L-rhamnopyransyl-fl-D- glucopyranoside and 3-cyanidin 6"-0-a—L-rhamnopyranosyl-,6-D—glucopyranoside (Wang et al., 1997) at 65 and 35%, respectively. Tumor Number and Size. The mice were sacrificed after 8 weeks of dietary treatment. A sample of liver tissue was obtained from each mouse for confirmation of Apc gene status by a PCR-based procedure (Su et al., 1992). The entire small 83 intestine, cecum, and colon were removed from each mouse. Intestinal sections were opened longitudinally and rinsed thoroughly with water. Sixteen ApcMin mice from each treatment were used for cDNA microarray analysis (8 males and 8 females). The middle and distal two-thirds of the small intestine were fixed overnight in 10% neutral buffered formalin (NBF) and then stained with 0.2% methylene blue. Tumor numbers and dimensions for the cecum and colon were determined immediately on unfixed tissue sections — these sections and proximal small intestine were then used for microarray analysis. For the remaining mice, all tissues were fixed in 10% NBF prior to tumor measurement and embedded for immunohistochemistry. Tumor numbers and dimensions for each intestinal segment were determined by direct counting with the aid of a dissecting microscope and measuring grid. The tumor sizes were determined by measuring the spherical (three dimensional) volume of adenomas in the cecum and colon and the average diameter of tumors in the small intestine. Tumors in the cecum and colon of ApcMin mice typically are polypoid in appearance, whereas the small intestinal tumors are seSSile. Spherical volumes of cecal and colonic tumors were calculated by the formula: Volume = 0.523 x (width x length x height of tumor). Weekly body weights were statistically analyzed by repeated-measures analysis of variance as a split-plot design. Main plot terms included treatment, sex, Apc status and their interactions. Sub-plot terms included time and its interactions with main plot effects. Tissue lengths were analyzed by two-way analysis of variance (AN OVA) to detect the effects and potential interactions of treatment and sex. Tumor numbers and diameters in the small intestine were analyzed by two-way 84 ANOVA (treatment, sex). For tumor numbers and volumes in cecum and colon, data were transformed to ranks and then ranks were analyzed by two-way AN OVA. When significant effects were detected (P<0.05), appropriate means were compared using the Least Significant Difference method. Mice which did not carry the APCMin gene mutation, and therefore did not develop intestinal tumors, were excluded from the final statistical analyses of tumor number and size. Microarray Analysis. RNA Isolation and Extraction Sixteen (8 males and 8 Min females) Apc mice from each treatment were used for microarray analysis. Total RNA was extracted from pooled samples (4 mice with same sex for each pooled sample) of the proximal small intestine, cecum and colon of these 16 ApcMin mice for each treatment. Tumors in the cecum and colon were quantified under a dissecting microscope and excised prior to epithelial cell isolation. Colon and cecum were scraped with a glass slide to obtain epithelial cells. RNAlaterTM (200 to 1000 111, Ambion, Austin, TX) was added and cells were stored at —80 °C. Approximately 800 mg of cells were obtained from the proximal small intestine section of each mouse, and approximately 200 mg and 250 mg of cells were obtained from the cecum and colon of each mouse, respectively. Equal amounts of cells from each mouse were pooled within same treatment, sex and tissue and used for RNA extraction and microarray analysis. The improved single step RNA isolation method (Chomczynski and Sacchi, 1987) was used to isolate total RNA from epithelial cells. RNA was prepared with Trizol LS reagent (Gibco BRL, Grand Island, NY) via manufacturer’s instructions. Briefly, cell pellets were resuspended in Trizol and incubated 5 minutes at room 85 temperature. Chloroform was added to the suspension to a final concentration of 16 % (V/V) and phases were separated by centrifugation for 5 minutes (10,000 x g). The aqueous phase was removed and RNA precipitated with isopropyl alcohol. The total RNA pellet was washed with 75% ethanol and concentration determined by spectrophotometry. RNA integrity was monitored by agarose gel electrophoresis. Fluorescence Labeling of Microarray Probes. F our pooled RNA samples for each tissue site (2 males and 2 females) were prepared from 8 male and 8 female ApcM’n mice for each treatment. Methods for fluorescence labeling followed the protocol developed by Hegde et al. (2000). cDNA was prepared by reverse transcription of total RNA samples (15 to 40 rig) in the presence of aminoallyl modified dUTP (aa- dUTP). Reverse transcription was performed with Superscript II (Gibco) via manufacturer’s instructions. Briefly, the secondary structure of RNA was relaxed in the presence of anchored oligo dT (T21VN). cDNA was produced upon addition of dNTPs (aa-dUTP: dTTP 2:1) and Superscript II (20 units) and incubation at 42 °C for 2 hours. Unincorporated dNTPs were removed using commercially available kit (Qiagen PCR, Qiagen Inc., Valencia, CA) and probes were evaporated in a speed vaccum for 1 hour. Dye coupling was done by incubation of cDNA with the monoreactive dyes cyanine 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 following addition of hydroxylamine at a final concentration of 2M. 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 86 resuspended in buffer (50% forrnamide, 5X SSC, 0.1% SDS + 20 pg COT-l DNA and 20 ug polyA DNA). Each probe mixture then was applied to microarray slides in a loop design (Fig. 19). After adding the probes, the slides were coverslipped and placed in a hybridization oven at 42 °C for 20 hr. The slides then were removed and washed with 1X SSC /0.2 % SDS until the cover slips were released. The slides were again washed two times with 0.1X SSC/0.2% SDS for 5 min and 0.1XSSC for 1 min to remove residual SDS. Preparation of the microarray slides. Microarray slides were printed with 15,360 genes received from the National Institute on Aging’s (NIA) mouse 15,264-gene (15K) set. The 15K cDNA clones were rearrayed from 52,374 ESTs (expressed sequence tags) derived from various embryonic and neonatal tissues. Full details on development on the 15K gene set are described by Tanaka et al. (2000). The 15K gene set was supplemented with an additional 96 known genes to allow symmetrical printing of genes in the slides. The cDNA library contains sequences coding for genes with known firnction, no hit genes (genes with no sequence information), unknown genes (genes with sequence information only), and genes coding for hypothetical protein (genes encoding a hypothetical protein based on high BLAST [basic local alignment search tool] score). Printing of this array was carried out using a superaldehyde substrate according to the manufacturer’s protocol (Telechem). Briefly, PCR amplification of inserts from each plasmid was performed using primers that contain a C6 amine group coupled to the 5’ end. The amplicons were precipitated and resuspended in spotting solution (3XSSC). The solutions were robotically spotted (Omnigrid robot, 87 GeneMachines, CA) onto superaldehyde coated glass slides at the Plant Research Laboratory at Michigan State University. The unbound DNA was removed and the bound probes denatured at 100°C. Free aldehydes were reduced with sodium borohydride and the slides washed extensively with 0.1X SSC/0.2% SDS and 0.1XSSC. Scanning, Immage Ouantitation, Normalization and Statistical Analysis. The slides were scanned in an array scanner (Affymetrix 428TM tray, Santa Clara, CA) and array image files were generated using the Jaguar 2.0 program TM (Affymetrix). Both Cy3 and Cy5 channels for each of the 15,360 genes (spots) on each slide were recognized, quantitated, and normalized against overall and peripheral background intensity level using the GenImage program developed by Dr. Sorin Draghici’s Bioinformatics lab at Wayne State University. These data were again normalized for dye variation using the GP3 R Script developed by Dr. Timothy Zachrawski’s lab at Michigan State University. A total of 36 pooled RNA samples were used in this analysis (2 male and 2 Mi" mice for each of the three treatments for each tissue female samples for Apc [small intestine, cecum, colon]). The full dataset, based on Cy3 and Cy5 channels in 72 slides, includes four independent estimates of expression for each of the 15,360 genes for each pooled RNA sample. Outlier observations were excluded by deleting any of the four independent observations of expression for each gene (for each RNA sample) that differed from the mean by more than one standard deviation. The remaining observations were averaged to obtain a single expression value for each gene for each RNA sample. These data were then statistically analyzed using SAS 88 (version 8.1) by tissue site and by gene using a 2-way analysis of variance (treatment, sex). When significant treatment effects were detected (P<0.05), mean values were compared using the Least Significant Difference method. 89 SI Colon Cecum //\\ /‘/’\\ //\\ A23 } A_—'S A—ES Figure 19. Aminoallyl labeling 100p of RNA for microarray analysis C, A and S represent three different dietary treatments, control, anthocyanins and sulindac, respectively. Each arrow represents a single microarray slide. In each slide the sample was labeled with Cy3 (tail of arrow) and also with Cy5 (head of arrow). The entire scheme was repeated four times, corresponding to the four pooled RNA samples (2 male, 2 female) obtained from animals on each treatment at each tissue site. (SI; small intestine) 90 B-catenin Immunohistochemistry Analysis. Expression of B-catenin in intestinal sections of normal and ApcMin mice exposed to the various dietary treatments was conducted. Four micrometer paraffin-embedded sections of small intestine, colon and cecum were deparaffinized and stained for B-catenin by indirect peroxidase biotin-streptavidin immunohistochemistry.. Sections were pretreated by an antigen retrieval procedure using 10 mM citrate buffer (pH 6) for 20 minutes at 95 °C and preblocked with 3% H202 in PBS followed by normal goat serum. After incubation of sections for 14 hr at 4 °C with anti-B-catenin monoclonal antibody (Transduction Laboratories, Lexington, KY) at a dilution of 1:250, biotinylated anti- mouse immunoglobulin (DAKO Laboratories, Carpinteria, CA) was applied to the sections at room temperature for 40 minutes. Sections then were treated with streptavidin-peroxidase (DAKO) at room temperature for 40 minutes. Linked peroxidase was Visualized by staining with aminoethylcarbazole (DAKO) for 30 minutes. The B-catenin immunostaining results for different locations within the tissue (top and bottom for small intestine and top, middle, and bottom for colon and cecum) and distribution within the cells (membrane and cytoplasm) were evaluated by visually comparing the staining intensities. The intensity of B-catenin immunostaining was scored using a scale ranging from absent (0) to very high (4). The scores for intensity of B-catenin immunostaining were transformed to ranks and then ranks were analyzed by four-way analysis of variance (treatment, Apc status, region within the tissue, and distribution within the cells). When significant 91 effects were detected (P<0.05), means were compared using the Least Significant Difference method. Images in this dissertation are presented in color. 92 RESULTS Anthocyanins Inhibit Intestinal Tumor Development. Averaged across all treatments, sexes and time points, mice consuming control (21.8 g) and anthocyanins (21.3 g) weighed more than mice consuming sulindac (20.3 g) (P<0.05). Male mice weighed significantly more (P<0.01) than female mice both at the beginning (14.0 versus 13.0 grams) and the end (26.4 versus 22.3 grams) of the experiment (Fig. 20). Final body weights of ApcMin mice consuming control diet, anthocyanins, and sulindac averaged 24.8, 23.5, and 24.9 grams , respectively. The lengths of the small intestine, cecum and colon were not influenced by dietary treatment (data not shown). Mice consuming sulindac had more (P<0.05) adenomas in the cecum than mice consuming control diet or anthocyanins (Fi g. 21). Cecal adenoma number for mice consuming control diet also was greater (P<0.05) than in mice consuming anthocyanins. The average number of adenomas in the cecum of mice that consumed anthocyanins, sulindac and control diet was 0.6, 2.1, and 1.0, respectively. Colonic adenoma number also was influenced by treatment (Fig. 21), with mice consuming control or sulindac having more tumors (P<0.05) than mice consuming anthocyanins. Average adenoma numbers in the colon were 0.8, 1.6, and 2.2 for mice that consumed anthocyanins, sulindac and control diet, respectively. The total burden (volume) of cecal adenomas was smallest (P<0.01) in mice consuming anthocyanins when compared to mice consuming control diet or sulindac (Fig. 22). 3 9 Adenoma volume in the cecum of mice that consumed anthocyanins was 0.7 mm whereas adenoma volumes in mice consuming control diet and sulindac were 1.5 and 4.7 m3, respectively (Fig. 22). The total burden of colonic adenomas was not 93 statistically significant at the P<0.05 level, but there was a trend that anthocyanins and sulindac reduced the total burden of colonic adenomas relative to mice consuming the control diet (P=0.08). Adenoma volumes in the colon of mice that 3 9 consumed anthocyanins, control, and sulindac were 2.6, 5.6, and 3.0 mm respectively (Fig. 22). Tumor multiplicity in the small intestine was significantly influenced by treatment (P<0.05). Adenoma number of the small intestine was obtained by combing adenoma numbers of the middle and distal small intestine segments. Mice that consumed sulindac had the smallest number of small intestinal adenomas (9 per mouse) whereas mice that consumed the control diet had the greatest number of , small intestinal adenomas (27 per mouse) (F i g. 23). Adenoma number of small intestine was intermediate in mice consuming anthocyanins (17 per mouse) (Fig. 23). Mice consuming sulindac had significantly fewer adenomas in both middle and distal small intestine segments than mice consuming control diet, and significantly fewer adenomas in the distal small intestine than mice consuming anthocyanins (P<0.05) (Fig. 23). The average size of small intestinal adenomas (Fig. 24) was reduced (P<0.05) by feeding sulindac (1.1 mm) relative to that observed in mice consuming the control diet (1.3 mm). The average size of small intestinal adenomas was intermediate in mice consuming anthocyanins (1.2 mm) (Fig. 24). 94 a) Male normal mice b) Female normal mice —*—C +3 ---A---A —x—C —-——S man-A 35 3o 304 .5" 251 3.421- 25“ IBM 20 m 1,. . E 20~ 4‘55 E” (U / (015- 6 15:1 6 10¢ 10" 5« 5: 0 I I ' ' I ' I 0 I r T T I r f o 1 2 3 4 5 6 7 8 o 1 2 3 4 s 6 7 8 VVeek VVeek c) Male ApcMm mice (1) Female ApcMm mice —x—C —I—S ---a---A 25 j 20’ fiafldfl Earn” a aw (U 15 (U {I L— L.- o 1 om 10« 5« 5‘ 0 1' T Y T Y I U 0 I U I I I I I o 1 2 3 4 s 6 7 8 o 1 2 3 4 5 5 7 8 Week Week Figure 20. Influence of treatment on body weights (g) of mice throughout the experimental period. C; control, S; sulindac, A; anthocyanins. Error bars indicate SEM. a) Body weights of male normal mice b) Body weights of female normal mice c) Body weights of male ApcMin mice (1) Body weights of female ApcMin mice 95 Cecum I Colon I" more: Adenoma Number 8 0.5 Control Anthocyanins Sulindac Figure 21. Influence of anthocyanins and sulindac on adenoma number in cecum (gray) and colon (black). Error bars indicate SEM. Columns of the same shade not sharing a common superscript are significantly different (P<0.05). 96 Cecum I Colon \) Adenoma Volume (mma) N w «h Uu ON fl 0 Control Anthocyanins Sulindac Figure 22. Influence of anthocyanins and sulindac on total adenoma volumes (mm3) in cecum (gray) and colon (black). Error bars indicate SEM. 8’ b’ cSignificant treatment effect (P<0.05). Columns not sharing a common superscript are significantly different. d' cTreatment effect trend (P=0.08). 97 EMSI DSI ITotal Adenoma Number Control Anthocyanins Sulindac Figure 23. Influence of anthocyanins and sulindac on adenoma number in small intestine. MSI; middle small intestine, DSI: distal small intestine. Error bars indicate SEM. Columns of the same shade not sharing a common superscript are significantly different (P<0.05). 98 .3 b 1 CT Adenoma Mean Diameter (mm) o oo Control Anthocyanins Sulindac Figure 24. Influence of anthocyanins and sulindac on mean diameters (mm) of adenomas in small intestine. Error bars indicate SEM. Columns not sharing a common superscript are significantly different (P<0.05). 99 Anthocyanins Do Not influence Intracellular Distribution of B-catenin. When the score of intensity of B-catenin immunostaining was compared, the expression of [3-catenin protein was consistently higher at the top of villi in the small intestine and top one third of crypts in cecum and colon of all carrier (ApcMin) and normal (Apc*’+) mice (P<0.05) (Fig. 25, 26 and 27). Within epithelial cells, expression of B-catenin protein was higher in cell membrane than in cytoplasm in all tissue sections (P<0.05) (Fig. 25, 26 and 27). Nuclear expression of B-catenin protein was negligible in all tissue sections. The expression of B-Catenin protein was Min greater (P<0.05) in intestinal tissues of Apc mice than those of normal mice in all sites (Fig. 25, 26 and 27). Mice consuming anthocyanins had increased expression of B-catenin protein in the cecum relative to mice consuming control diet or sulindac (P<0.05) (Table 4). Significant interactions between treatment and Apc status were found in the proximal and middle small intestinal sections. The expression of B-catenin protein was increased in the proximal small intestine as well as in the middle small intestine of Min Apc mice consuming anthocyanins compared with those consuming control diet (P<0.05) (Table 4). 100 (a) Small Intestine N (Apc+’*) C (Ape+ '“) (b) Colon N (Apc*’*) C (Ape+ i") (c) Cecum N (Apc+ +) C (AchMm) Figure 25. Expression of B-catenin in mice consuming the control diet. 101 (a) Small Intestine wah qmfl" (b) Colon NMWD CNW”D (c) Cecum N (Apc+ 4») C (AchMin) Figure 26. Expression of [El-catenin in mice consuming sulindac 102 (a) Small Intestine wipe“) " I V C(Ap") (b) Colon N (APCH) C (Apc* (c) Cecum ’ N (Ape+ ‘) C (Ape+ " I.) Figure 27. Expression of B-catenin in mice consuming anthocyanins 103 Table 3. Average intensity scores of B-catenin expression in intestinal sectionsl’ A. Membrane staining in normal mice 2 Treatment PSI3 MSI3 DSI Colon Cecum4 Control 1.8 i- 0.3abc 1.5 i- 0.2b 1.5 i 0.4 1.7: 0.4 1.2 i- 0.221 Sulindac 1.5 i 0.3ab 1.3 3. 0.2“ 2.3 i 0.3 1.2 i 0.3 1.3 i 0.38 . Anthocyanins 1.5 i 0.33 2.3 i 0.2c 2.0 i 0.3 1.8 i 0.3 2.0 i 0.2b B. CytoSol staining in normal mice Treatment PSI MSI DSI Colon Cecum Control 0.7 i 0.3“bc 0.3 i 0.2b 0.5 i 0.4 0.7 i 0.4 0.7 i 0.23 Sulindac 0.5 :1: 0.3ab 0.3 i 0.2“b 0.8 i 0.3 0.7 i 0.3 0.3 i 0.3a Anthocyanins 0.3 i 0.3a 0.8 :r. 0.2c 0.5 i 0.3 0.7 i 0.3 0.8 i 0.2b C. Membrane staining in ApcMin mice Treatment PSI MSI DSI Colon Cecum Control 1.5 i 0.2“ 0.8 i 0.2“ 1.8 3. 0.2 1.8 i 0.2 1.6 i 0.28 Sulindac 1.7 i 0.2be 1.5 i 0.2b 2.3 i 0.2 2.1 i 0.2 1.4 i 0.23 Anthocyanins 2.0 i 0.2c 2.3 i 02° 2.3 i 0.2 2.0 i- 0.2 2.1 i 0.1b D. Cytosol staining in ApcMin mice Treatment PSI MSI DSI Colon Cecum Control 0.3 i 02“" 0.0 i‘ 0.23 0.5 i- 0.2 0.9 i 0.2 0.6 i 0.23 Sulindac 0.8 _+. 0.2be 0.5 i 0.21) 0.7 :1: 0.2 0.9 i 0.2 0.3 i 0.2a Anthocyanins 1.0 i 02° 1.3 i 02° 0.8 :1: 0.2 1.4 i 0.2 0.8 i 0.2b IPSI; proximal small intestine, MSI; middle small intestine, DSI; distal small intestine. 2The intensity of B-catenin immunostaining was scored using a scale ranging from absent (0) to very high (4). 3Treatment x carrier status interaction (P<0.05). Means in the same column not sharing a common superscript are significantly different (p<0.05). 4Treatment effect (P<0.05). Means in the same column within each category not sharing a common superscript are significantly different (p<0.05). Anthocyanins and Sulindac Do Not Influence Expression of mRNA of APC, B-catenin and Most of the Predicted Target Genes of Wnt/B-catenin Signaling. Expression of B-catenin and the predicted target genes of the Wnt signaling pathway was determined in small intestine, colon and cecum of ApcMin mice using cDNA microarray analysis (Table 4). Anthocyanins and sulindac did not alter the expression of mRNA of APC and B-catenin in all three intestinal sections of ApcMin mice. For the predicted target genes of the Wnt/B-catenin signaling, anthocyanins decreased the expression of cyclin D1 compared to sulindac in cecum (0.59 vs 0.79). Treatment did not influence the expression of mRNA for the other predicted target genes — MMP7, PPARB and C-Myc - in all three tissue sections of ApcMin mice. cDNA Microarray Analysis Identified Several Clusters of Differentially Expressed Genes in Small intestine, Colon and Cecum of ApcMin Mice. All gene identifications were based on BLAST scores greater than 100 as of May 1, 2002. There were 497 genes influenced by any treatment (F value for treatment effect < 0.05) in SI, 534 in Colon, and 501 in cecum (Fig. 28). None of the genes was influenced by treatment in all three intestinal sections. There were 8 genes influenced by treatment both in small intestine and colon (Table 5). Relative to controls, sulindac down-regulated 3 of these 8 genes in small intestine and four genes in colon. Sulindac up-regulated 2 genes in colon compared with controls. Relative to controls, anthocyanins up-regulated 2 Of the 8 genes and down-regulated three genes in small intestine. Anthocyanins down-regulated three of the 8 genes in colon compared with controls. There was little overlap in the effects of sulindac and 105 anthOcyanins on expression of these eight genes. In small intestine, both compounds down-regulated the expression of a single gene (unknown function). Both compounds down-regulated the expression of placental lacto gen 2 in colon, but expression of this gene was up-regulated by anthocyanins and not influenced by sulindac in small intestine. There were 12 genes influenced by treatment both in small intestine and cecum (Table 6). These include 6 genes coding for known proteins (voltage- dependent calcium channel beta-3 subunit, grth arrest specific protein 5, tubulin beta 3, collagen IV, kangai), 5 unknown and 1 hypothetical protein gene. Of these 12 genes, sulindac reduced expression of 3 genes in small intestine and none of the genes in the colon when compared to controls. None of these 12 genes were up- _ regulated by sulindac relative to control. In small intestine relative to controls, anthocyanins reduced the expression of calcium channel beta 3 subunit, growth arrest protein 5, tubulin beta 3, collagen IV and one unknown gene and increased the expression of alternative splicing factor, 1 unknown gene, and 1 hypothetical protein. Expression of these genes was similary affected by anthocyanins in cecum except for an increased expression of calcium channel subunit and decreased expression of kangai 1, (Table 6). Twenty genes were differentially influenced by treatment in both cecum and colon. Among those genes changed both in colon and cecum, anthocyanins up- regulated 9 genes and down-regulated 3 genes compared to both controls and sulindac in cecum (Table 7). The up-regulated genes were nuclear antigen SplOO, serine/threonine-protein kinase 4, acid ceramidase 1, CD36 antigen-like 2, signal 106 transducing adaptor molecule 2, UMP-CMP kinase, and 3 unknown genes. The down- regulated genes relative to both control and sulindac were zinc finger protein RIN ZF, 1 no hit, and 1 hypothetical protein gene. In colon, sulindac influenced the expression of most of these 20 genes, whereas anthocyanins did not, compared to control. Relative to controls, sulindac up-regulated genes for zinc finger protein LIN ZF, MORF-related gene X, nuclear antigen SplOO, serine/threonin-protein kinase 4, acid ceramidasel, 1 no hit, and 7 unknown genes and down-regulated CD36 anti gen- like 2, signal transducing adaptor molecule 2, UMP-CMP kinase, 1 hypothetical protein, and 1 no hit gene (Table 7). Differentially Expressed Genes in Cecum of ApcMin Mice. Cecal - epithelium from anthocyanin-treated mice had significantly increased expression of 79 genes compared to mice fed the control and sulindac diets (Table 8). Anthocyanins increased the expression of the hemachromatosis (HFE) gene, ras- related C3 botulinum toxin substrate 1, and Ly6/Neurotoxin 1 protein genes more than 2-fold compared to both control and sulindac. Anthocyanins also significantly decreased the expression of 44 genes compared to both control and sulindac (Table 9). The decrease in expression of 2 unknown genes by anthocyanins was more than 2-fold compared to control or sulindac. Differentially Expressed Genes in Colon of ApcM’" Mice. Anthocyanins significantly increased the expression of 33 genes in colonic epithelium compared to both control and sulindac treated mice (Table 10). Anthocyanins significantly decreased the expression of 33 genes compared to both control and sulindac treated 107 mice (Table 11). The larger fold changes were an increase of expression for cytochrome b5, adolase C5 and 1 unknown gene (Table 10) and decrease for serum protein MSE55 (Table 11). Differentially Expressed Genes in Small Intestine of ApcMin Mice. In ' small intestine, 53 and 244 genes were up-regulated by sulindac (Table 12 and 13) and anthocyanins (Table 13 and data not shown), respectively, when compared to controls. Among these genes, 19 genes were up-regulated by sulindac and anthocyanins (Table 13). The genes that were increased in their expression in small intestine both by sulindac and anthocyanins include 9 known genes. Among genes influenced specifically by sulindac (Table 12), the expression of ATPase subunit 4 gene was increased by 3.36-fold in mice consuming sulindac compared to mice consuming the control diet. The expression of Tax interacting protein gene was increased most (1.5-fold) by sulindac and LIM domain binding 1 gene (1.79-fold) by anthocyanins (Table 13). Sulindac and anthocyanins also down-regulated a large group of genes in the small intestine. 108 genes were down-regulated by sulindac (Table 14 and 15) and 47 genes were down-regulated both by sulindac and anthocyanins (Table 15) when compared to control. Genes that were down-regulated specifically by sulindac more than 1.5-fold were 1 no hit and 1 unknown gene. Adipocyte-specific protein 5 and novel cell death-regulatory protein GRIM19 were down-regulated by 1.49-fold and 1.44-fold, respectively (Table 14). Genes that were down-regulated more that 1.5- fold in their expression by both sulindac and anthocyanins were fatty acid binding protein 3, glutathione S-transferase and 2 unknown genes (Table 15). 108 Table 4. Expression of B-cate'nin and target genes of Wnt/B-catenin signaling in intestinal sections of ApcMin mice" 2 Plate ID3 Gene Name C S A SEM4 Small Intestine H3080G10 APC 0.75 0.69 0.77 0.08 H3080F09 APC 0.62 0.71 0.63 0.05 H3031E05 B-catenin 1.26 1.59 1.25 0.23 H3084D05 Cyclin D1 0.67 0.64 0.57 0.04 H3093H12 MMP7 0.69 0.71 0.80 0.04 H3155B12 PPARdelta 1.48 1.13 1.45 0.24 H3 076D1 0 C-myc 1.26 1.27 1.17 0.12 H3089H11 C-myc 0.87 0.80 0.70 0.09 Colon H3080G10 APC 0.64 0.76 0.72 0.07 H3080F09 APC 0.73 0.78 0.79 0.04 H3031E05 B-catenin 1.45 1.54 1.67 0.20 H3084D05 Cyclin D1 0.66 0.61 0.62 0.06 H3093H12 MMP7 0.84 0.83 0.79 0.05 H3155B12 PPARdelta 1.16 1.08 1.18 0.14 ' H3076D10 C-myc 1.24 0.97 1.00 0.1 1 H3089H11 C-myc 0.63 0.71 0.67 0.06 Cecum H3080G10 APC 0.68 0.73 0.82 0.06 H3080F09 APC 0.72 0.69 0.68 0.03 H3031E05 B-catenin 1.48 1.70 1.67 0.31 H3084D05 Cyclin D1 0.69ab 0.76b 0.59a 0.16 H3093H12 MMP7 0.88 0.85 0.70 0.12 H3155812 PPARdelta 1.18 1.09 1.16 0.27 H3076D10 C-myc 1.01 0.93 0.90 0.14 H3089H11 C-myc 0.64 0.65 0.71 0.06 IData represent mean intensity values of mRNA expression for the corresponding gene in the treatment groups, C(control), S(sulindac) and A(anthocyanins). Microarray slides have two spots for a few identical genes. APC and C-myc have two spots each and 2 intensity values from each spot were obtained. 3 Plate ID: sequence number for NIA 15K mouse cDNA library set. 4SEM; standard error of mean. a""cMeans in the same row not sharing a common superscript are significantly different (P<0.05). 109 Small Intestine Figure 28. Numbers of genes influenced by any treatment (P<0.05) in intestinal sections of APCMin mice. Numbers overlapping two sections were influenced by treatment in both tissues. 110 Table 5. Expression of 8 genes influenced by treatment both in small intestine and colon1 Plate ID2 Gene Name C S A SEM3 Small Intestine H3046BO4 placental lactogen2 0.66a 0.753 0.94b 0.05 H3085E07 unknown 0.72a 0.85ab 0.91b 0.04 H3022F05 3-phosphoglycerate dehydrogenase (Phgdh) 0.84ab 0.7821 1.06b 0.06 H3084BO6 F-box protein FBX15 . 0.80b 0.65a 0.88b 0.03 H3045C02 ubiquitin-conjugating enzyme 0.69b 0.54a 0.71b 0.02 H3003D111ymphoid-specific transcription factor NFATc3 1.70b 1.34ab 0.71a 0.20 H3090D11unknown 0.89b 0.708 0.71“ 0.04 H3015G01no hit found 1.611) 1.27““’1.16a 0.10 Colon H3046B04placentallactogen2 0.80c 0.72b 0.64a 0.01 H3085E07 unknown 0.66a 0.93b 0.63“ 0.06 H3022F05 3-phosphoglycerate dehydrogenase (Phgdh) 0.75a 0.88b 0.82ab 0.03 H3084B06 F-box protein FBX15 0.84b 0.67b 0.5921 0.06 H3045C02 ubiquitin-conjugating enzyme 0.73b 0.75b 0.67a 0.02 H3003D111ymphoid-specific transcription factor NFATo31.92b 1.32a 1.71b 0.09 H3090D11unknown 0.92b 0.71a 0.92"~0.04 H3015G01no hit found 1.48b 1.2521 1.59b 0.05 rData represent mean intensity values of mRNA expression for the corresponding gene in the treatment groups, C(control), S(sulindac) and A(anthocyanins). Plate ID: sequence number for NIA 15K mouse cDNA library set 3SEM; standard error of mean. 2|“Means in the same row not sharing a common superscript are significantly different (P<0.05). Ill Table 6. Expression of 12 genes influenced by treatment both in small intestine and l cecum Plate ID? Gene Name C S A SEM3 Small Intestine voltage-dependent H3117A06 calcium channel beta-3 subunit 0.75“ 0.59“ 0.64“ 0.02 H3113A12 growth arrest specific protein5 1.07“ 0.88““ 0.79“ 0.06 H3057B12 tubulinbeta3 1.44“ 1.27““ 1.00“ 0.08 H3067F05 collagenIV 0.96“ 0.72“ 0.66“ 0.06 H3051G02 unknown 1.68“ 1.46“b 1.07“ 0.13 kangai 1 H3154D02 (suppression of tumorigenicity 6) 0.82ab 0.61a 0.91b 0.06 H3060C12 unknown 0.84b 0.66“ 0.78“ 0.03 H3119E07 hypothetical 0.75“ 0.73“ 0.86b 0.03 H3104H02 alternative splicing factor 0.79a 0.813 0.98b 0.02 H3033B06 unknown 0.74““ 0.65“ 0.94“ 0.05 H3042A04 unknown 0.74“ 0.69“ 0.87“ 0.03 H3064D02 unknown 0.73““ 0.63“ 0.80“ 0.03 Cecum voltage-dependent H3117A06 calcium channel beta-3 subunit 0.62“ 0.61“ 0.75“ 0.03 H3113A12 growth arrest specific protein5 1.78“ 1.55““ 1.38“ 0.08 H3057B12 tubulin beta3 3.16“ 2.40““ 1.94“ 0.26 H3067F05 collagen 1v 0.88“ 0.91“ 0.66“ 0.05 H3051G02 unknown 1.35“ 1.16““ 0.81“ 0.12 kangail H3154D02 (suppression oftumorigenicity 6) 0.84“ 0.70““ 0.63“ 0.04 H3060C12 unknown 0.62“ 0.63“ 0.73“ 0.02 H3119E07 hypothetical 0.63“ 0.62“ 0.98“ 0.04 H3104H02 alternative splicing factor 1.55“ 1.29“ 1.04“ 0.08 H3033B06 unknown 0.68“ 0.67“ 0.91“ 0.05 H3042A04 unknown 0.77“ 0.76b 0.62“ 0.04 H3064D02 unknown 0.61“ 0.59“ 0.73“ 0.02 TData represent mean intensity values of mRNA expression for the corresponding gene in the treatment groups, C(control), S(sulindac) and A(anthocyanins). Plate ID: sequence number for NIA 15K mouse cDNA library set 3SEM; standard error of mean. a'b'cMeans in the same row not sharing a common superscript are significantly different (P<0.05). 112 Table 7. Expression of 20 genes influenced by treatment both in cecum and colon1 Plate ID2 Gene Name C S A SEM3 Cecum H3112C06no hit found 3l.02“30.29“7.06“ 4.99 histocompatibility complex region H3073B01containingtheQregion ofclassI 0.76“ 0.69““ 0.59“ 0.04 H3061A052inc finger proteinRINZF (RINZF) 0.82b 0.85b 0.663' 0.04 H3114G07no hit found 1.25“ 1.04“ 1.01“ 0.05 H3117D10hypothetical 0.93“ 0.87b 0.78“ 0.03 H3088E03 unknown 0.85““088“ 0.77“ 0.03 H3132A08unknown 0.64““ 0.70“ 0.59“ 0.02 H3148F01 unknown 0.69“ 0.60“ 0.66““ 0.02 H3068E08 unknown 0.66“ 0.81“ 0.63“ 0.03 H3030E03 unknown 0.66“ 0.77“ 0.71““ 0.02 H3078H05MORF-related geneX 0.70““065“ 0.83“ 0.04 H3052B09nuclear antigen Sp100 (Sp100) 0.61" 0.612’ 0.74b 0.03 H3056H07unknown 0.59“ 0.63“ 0.72“ 0.02 SERINE/THREONINE-PROTEIN KINASE 4 H3138A02(P21-ACTIVATED KINASE 4) 0.58“ 0.61“ 0.74“ 0.02 H3092C04 acid ceramidase(Asah1) 0.57“ 0.64“ 0.78“ 0.04 H3156B09 unknown 0.63“ 0.63“ 0.90“ 0.03 CD36 antigen (collagen type I receptor, H3111D12thrombospondin receptor)-like2(Cd3612) 1.74a 1.858 2.53b 0.19 signal transducing adaptor molecule H3007H10(SH3 domain and ITAM motif)2 1.76“ 1.51“ 2.79“ 0.28 H3144A09unknown 0.83“ 0.82“ 1.35“ 0.13 H3007F10 UMP-CMP kinase (UMP-CMPK) 1.51“ 1.62“ 2.69“ 0.16 Colon H3112C06no hit found 25.10“7.04“ 22.14“3.29 histocompatibility complex region H3073BOl containing theQregion ofclassI 0.69““ 0.79“ 0.64“ 0.03 H3061A052inc finger proteinRINZF (RINZF) 0.69a 0.90b 0.68‘1 0.05 H3114G07no hit found 1.08“ 1.61“ 1.20“ 0.06 H3117D10hypothetical 0.97“ 0.75“ 0.79“ 0.04 H3088E03 unknown 0.71“ 0.85“ 0.75“ 0.02 H3132A08unknown 0.62“ 0.79“ 0.68“ 0.03 H3148F01 unknown 0.62“ 0.72“ 0.68““ 0.02 H3068E08 unknown 0.59“ 0.72“ 0.65“ 0.02 113 H3030E03 unknown 0.69“ 0.94“ 0.62“ 0.04 H3078H05MORF-related geneX 0.70“ 0.88“ 0.68“ 0.05 H3052B09nuclear antigen Sp100(Sp100) 0.59“ 0.73“ 0.63““ 0.03 H3056H07unknown 0.66““ 0.72“ 0.58“ 0.03 SERINE/THREONINE-PROTEN KINASE 4 H3138A02(P21-ACTIVATED KINASE 4) 0.65“ 0.85“ 0.66“ 0.05 H3092C04 acid ceramidase(Asah1) 0.63“ 0.82“ 0.62“ 0.04 H3156B09unknown 0.81“ 0.89“ 0.81“ 0.02 CD36 antigen (collagen type I receptor, . H3111D12thrombospondin receptor)-like 2 (Cd3612) 2.01b 1.40a 1.87b 0.13 signal transducing adaptor molecule H3007H10(SH3 domain and ITAM motif)2 2.67“ 1.79“ 2.51“ 0.19 H3144A09unknown 0.67“ 0.82“ 0.85“ 0.03 H3007F10 UMP-CMP kinase (UMP-CMPK) 2.92“ 1.76“ 2.86“ 0.27 IData represent mean intensity values of mRNA expression for the corresponding gene in the treatment groups, C(control), S(sulindac) and A(anthocyanins). Plate ID: sequence number for NIA 15K mouse cDNA library set 3SEM; standard error of mean. a’b‘cMeans in the same row not sharing a common superscript are significantly different (P<0.05). 114 Table 8. List of genes up—regulated by anthocyanins vs control and sulindac in cecum Plate IDI Gene Name2 A/C3 A/S3 H3144FO8 hemochromatosis (HFE) gene 2.86 3.26 ras-related C3 botulinum toxin substrate 1 H3018C09 (rho family, small GTP binding protein Rac 1) 2.64 2.79 H3131F08 Ly6/neurotoxin 1 protein 2.33 3.38 H3004AO9 unknown 1.79 1.61 H3042E10 unknown 1.75 1.60 H3001H08 adrenomedullin precursor 1.71 1.79 H3101B09 unknown 1.61 1.67 H3016E09 unknown 1.59 1.42 H3144C09 tetratricopeptide repeat domain 1.58 1.49 H3119E07 hypothetical 1.55 1.57 H3004B09 mitochondrial ribosomal protein S7 1.55 1.39 H3078D06 unknown 1.49 1.48 H3004F11 unknown 1.47 1.52 H3014HO2 hypothetical 1.45 1.38 H3023DO9 unknown 1.45 1.42 H3045D09 NEUROENDOCRINE DIFFERENTIATION FACTOR 1.45 1.35 H3051H08 initiate factor 5 1.44 1.55 H3061E10 unknown 1.44 1.35 H3104E01 unknown 1.44 1.46 H3146B05 unknown 1.44 1.25 H3035612 unknown 1.44 1.45 H3012B09 nemo-like kinase 1.43 1.33 H3006D05 no hit found 1.41 1.31 H3023C01 hypothetical 1.41 1.39 H3014GO7 astrocytic phosphoprotein 1.40 1.36 H3008G10 bladder cancer associated protein 1.39 1.44 H3008G05 translocase of inner mitochondrial membrane 8 1.39 1.36 H3001A02 secretin 1.37 1.41 H3094G12 unknown 1.35 1.26 H3082F05 unknown 1.34 1.36 H3144GO9 unknown 1.33 1.26 H3047H09 unknown 1.32 1.17 H3033B06 no hit found 1.32 1.35 H3067GO9 unknown 1.32 1.27 H3067GlO unknown 1.29 1.25 H3098E01 no hit found 1.29 1.25 H3052C306 p1astin2 1.29 1.25 H3064F09 chloride channel regulator Icln pseudogene 1.28 1.26 H3147FO6 malate NADP oxidoreductase 1.28 1.23 115 H3081EO7 H3082A04 H3100F05 H3 047C03 H3134C08 H3098802 H3’06OC06 H3035E03 H3039A12 H3134A09 H3OSSBO7 H3027AO7 H3060D05 H3080E02 H3 087C08 H3022DO9 H3 O37AO4 H3134E02 H3052C08 H3014B04 H3082F09 H31 17AO6 H3065G09 H3064D02 H3157BO9 H3044E02 H3045C06 H3060C12 H3084CO3 H3134H09 H3065E12 H3074E09 H3032A02 H3133HO4 H3055A04 H3093G12 H3126A12 H3057E08 H3027D1 1 H3054F10 Sjogren syndrome antigen A2 unknown unknown early onset breast cancer protein 2 unknown unknown unknown hypothetical unknown unknown unknown translation repressor NATl beta-defensin 8 unknown unknown beta-glucoronidase no hit found solitary transfer RNA-Asp coproporphyrinogen oxidase unknown unknown voltage-dependent calcium channel beta-3 subunit unknown unknown unknown ribonucleotide reductase M2 unknown unknown hippocalcin—like 1 unknown unknown unknown unknown unknown unknown unknown DNA POLYMERASE EPSILON P12 SUBUNIT unknown unknown unknown 1.28 1.28 1.27 1.27 1.26 1.26 1.26 1.25 1.25 1.25 1.24 1.24 1.23 1.23 1.23 1.22 1.21 1.21 1.21 1.21 1.21 1.20 1.20 1.19 1.19 1.19 1.18 1.18 1.18 1.17 1.16 1.16 1.15 1.15 1.13 1.12 1.11 1.11 1.10 1.08 1.29 1.28 1.13 1.16 1.13 1.22 1.22 1.20 1.26 1.23 1.24 1.16 1.17 1.28 1.22 1.21 1.17 1.12 1.17 1.21 1.23 1.23 1.13 1.24 1.12 1.23 1.13 1.16 1.19 1.15 1.14 1.12 1.20 1.17 1.18 1.15 1.14 1.17 1.12 1.19 rPlate identification number: sequence number for NIA 15K mouse cDNA library set 2This list of genes includes those that were significantly up-regulated by anthocyanins compared to control and sulindac (P<0.05). Names written in capital 116 letters are for the respective homolog of the genes. The total number of genes that were up-regulated by anthocyanins is 79, which includes 44 unknown, 4 no hit found and 4 hypothetical protein genes. 3 Ratio of intensity score: A/C; anthocyanins versus control, A/S; anthocyanins versus sulindac. Ratio represents fold change in gene expression in tissue of mice consuming anthocyanins when compared that of mice consuming control or sulindac. 117 Table 9. List of genes down-regulated by anthocyanins vs control and sulindac in cecum Plate ID1 Gene Name2 C/A3 S/A3 H3119A02 unknown 2.27 1.77 H3146GO9 unknown 2.01 2.35 H3065C02 GTP-binding protein NGB 1.78 1.42 H3119C02 olfactory receptor gene cluster 1.72 1.71 H3003GO4 cell adhesion molecule nectin-3 alpha 1.63 1.55 H3104D01 beta-1,4-galactosyltransferase 1.57 1.45 H3039F01 LEUKOTRIENE B4 lZ-HYDROXYDEHYDROGENASE 1.55 1.51 H3117H05 OXYSTEROL BINDING PROTEIN-RELATED PROTEIN 3 1.55 1.43 H3072C07 unknown 1.45 1.28 H3157E05 zinc transporter 4 1.42 1.35 H3071EO4 unknown 1.37 1.44 H307 5A02 omithine decarboxylase 1.36 1.33 H3134D02 intermediate filament proteins family 1.36 1.32 H3 090H01 chromatin structural protein 1.34 1.33 H3094A05 hypothetical 1.34 1.28 H3035A05 no hit found 1.34 1.35 H3157D05 unknown 1.32 1.40 H3067F05 unknown 1.32 1.38 H3032FO4 unknown 1.31 1.26 H3082FO4 unknown 1.29 1.30 H3105F07 hypothetical 1.28 1.30 H3090D06 unknown 1.28 1.21 H3142G03 unknown 1.25 1.37 H3131EO6 AS oncogene family 1.25 1.25 H3042A04 unknown 1.24 1.23 H3044D08 unknown 1.24 1.25 H3131E05 unknown 1.24 1.20 H3074C05 unknown 1.24 1.16 H3081D02 Bc1-2-related ovarian killer protein, apoptosis activator 1.23 1.26 H3096C02 no hit found 1.22 1.29 H3033AO9 no hit found 1.21 1.33 H3067G06 no hit found 1.21 1.26 H3033C10unknown 1.19 1.19 H3044D03 unknown 1.19 1.16 H3072C05 unknown 1.19 1.12 H3150H07no hit found 1.18 1.14 H3105G09 aryl hydrocarbon receptor nuclear translocator-like 1.18 1.26 H3033D05 unknown 1.17 1.20 H3037C05 unknown 1.15 1.17 118 H3080607 unknown 1.15 1.14 H3117H06unknown 1.14 1.19 H3084A06spindlin 1.14 1.18 H3126D02unknown 1.14 1.12 H3136G08nohit found 1.12 1.11 IPlate identification number: sequence number for NIA 15K mouse cDNA library set 2This list of genes includes those that were significantly down-regulated by anthocyanins compared to control and sulindac (P<0.05). The total number of genes that were down-regulated by anthocyanins is 44, which includes 24 unknown, 6 no hit found and 2 hypothetical protein genes. 3Ratio of intensity score: C/A; control versus anthocyanins, S/A; sulindac versus anthocyanins. Ratio represents fold change in gene expression in tissue of mice consuming anthocyanins when compared that of mice consuming control or sulindac. 119 Table 10. List of genes up-regulated by anthocyanins vs control and sulindac in colon Plate IDl Gene Name2 A/C3 A/S3 H3029A08 cytochrome b5 1.73 1.36 H3063C01 unknown 1.53 1.54 H3051C03 aldolase C 1.52 1.48 H3009C07 heat shock protein 1.49 1.32 H3071C08 E(y)2 PROTEIN, CG10347 PROTEIN 1.48 1.37 H3069H09 unknown 1.44 1.31 H3008E10 no hit found 1.41 1.50 H3095E02 unknown 1.40 1.22 H3008C06 unknown 1.38 1.34 H3003G03 458 pre rRNA 1.37 1.36 H3075E12 TRANSPORTlN-SR 1.36 1.32 H3125GO9 unknown 1.34 1.25 H3036GO7 epithelial cell transmembrane protein antigen precursor 1.33 1.43 TRANSLOCON-ASSOCIATED PROTEIN, H3131A08 SIGNAL SEQUENCE RECEPTOR BETA SUBUNIT) 1.32 1.18 H3074A08 TRANSFORMING PROTEIN RHOA (RhoA GTPase) 1.32 1.35 H3051E03 unknown 1.31 1.48 H3042EO8 interleukin 1 receptor-associated kinase(111rak) 1.30 1.35 H3003DO6 inhibitor of growth family member 1 (INGl) 1.29 1.27 H3094GO6 unknown 1.27 1.37 H3108A08BIMP1 (apoptosis regulator) 1.27 1.34 H3136F07 guanine monphosphate synthetase (GMPS) 1.25 1.13 H3153H04 CALCINEURIN B 1.25 1.28 H3058C10 coagulation factor H receptor-like 3 (F2RL3) 1.23 1.34 H3010D11unknown 1.23 1.55 H3113G05unknown 1.21 1.16 H3122A12 S-adenosylmethionine decarboxylase 1.21 1.10 H3032A06butyrophilin-like 2 1.20 1.28 H3041G12 CELL CYCLE PROGRESSION RESTORATION GENE 3 1.15 1.11 H3044FO3 unknown 1.14 1.14 H3050C12 unknown 1.14 1.10 H3008810 ATPase, H+ transporting, lysosomal I (Atp6i) 1.14 1.26 H3008G04 GATA binding protein 2 1.13 1.23 H3006F08 unknown 1.13 1.34 TPlate identification number: sequence number for NIA 15K mouse cDNA library set 2This list of genes includes those that were significantly up-regulated by anthocyanins compared to control and sulindac (P<0.05). The total number of genes that were up-regulated by anthocyanins is 33, which includes 10 unknown genes and 1 no hit gene. 120 3Ratio of intensity score: A/C; anthocyanins versus control, A/S; anthocyanins versus sulindac. Ratio represents fold change in gene expression in tissue of mice consuming anthocyanins when compared that of mice consuming control or sulindac. 121 Table 11. List of genes down-regulated by anthocyanins vs control and sulindac in colon (I Plate 11)1 Gene Name2 C/A3 S/A3 H3130F06 SERUM PROTEIN MSE55 1.59 1.43 H3086B08 unknown 1.39 1.24 H3073E04 initiation factor 5 1.36 1.34 H3089F08 unknown 1.33 1.24 H3074H01 unknown 1.31 1.28 H304ODO3 PSD-95/SAP90-associated protein-4 1.30 1.35 H3123D04 unknown 1.29 1.29 H3089D05 hypothetical 1.28 1.44 H3026H05 unknown 1.27 1.36 H3053A06 inositol 1,4,5-trisphosphate 3-kinase B (ITPKB) 1.26 1.19 H3028H05 KLAAO938 protein (KIAAO938) 1.26 1.39 H3046B04 placental lactogen 2 1.25 1,14 PHOSPHOLYSINE PHOSPHOHISTIDINE H3105D11 INORGANIC PYROPHOSPHATE PHOSPHATASE 1.25 1.18 H3086H05 Deleted in colorectal cancer(Dcc) 1.24 1.34 H3026D05 N-myristoyltransferase 1 1.22 1.21 H3033FO6 neuronal apoptosis inhibitory protein 7 (Naip7) 1.21 1.15 H3017AO7 A kinase (PRKA) anchor protein (yotiao) 9 (AKAP9) 1.21 1.20 H3 O68H04 ribonuclease/angiogenin inhibitor 2 (Rnh2) 1 .21 1 . l 9 H3106BO7 orphan nuclear receptor (NR2E1) 1.20 1.20 H3090HO8 KIAAO676 PROTEIN 1.20 1.19 H3128F 02 hypothetical ' 1.19 1.26 H3045H05 unknown 1.19 1.30 H3068F08 protein tyrosine phosphatase, non-receptor type 21 1.19 1.25 H3096F11 HNK-l sulfotransferase 1.18 1.22 H3078H10 ADP-ribosylation factor 1 1.18 1.27 H3112F03 degenerative sperrnatocyte 1.18 1.21 H3056D05 no hit found 1.15 1.14 H3100D11no hit found 1.14 1.09 H3134CO9 Rhesus blood group protein (Rh) 1.14 1.33 H3068BOZ hypothetical 1.13 1.18 FYVE-FINGER-CONTAINING H3074F09 RABS EFFECTOR PROTEIN RABENOSYN-S 1.13 1.14 H3128811 putative oncogene (HLC14-06-P) 1.13 1.07 H3045C02 ubiquitin-conj ugating enzyme 1.09 1.1 1 IPlate identification number: sequence number for NIA 15K mouse cDNA library set 2This list of genes includes those that were significantly down-regulated by anthocyanins compared to control and sulindac (P<0.05). The total number of genes 122 that were down-regulated by anthocyanins is 33, which includes 6 unknown, 2 no hit and 3 hypothetical protein genes. 2Ratio of intensity score: C/A; control versus anthocyanins, S/A; sulindac versus anthocyanins. Ratio represents fold change in gene expression in tissue of mice consuming anthocyanins when compared that of mice consuming control or sulindac. 123 Table 12. List of genes up-regulated by sulindac vs control in small intestine Plate IDl Gene Name1r S/C3 H3111B05 no hit found 3.53 H3139D09 ATPase subunit 6 3.36 H3119B08 no hit found 2.43 H3016H08 no hit found 2.15 H3112H02 no hit found 2.06 H3020A01 no hit found 1.95 H3155HO7 unknown 1.85 H3117HO8 actin related protein 2/3 complex, subunit 4 1.77 H3 009D08 unknown 1 .72 H3109B12 putative nuclear protein 1.57 H3 069D02 hypothetical 1 .56 H3117C01 KRUPPEL—RELATED ZINC FINGER PROTEIN 1.55 H3114G09 no hit found 154 H3118A01 no hit found 1.52 H3 05 8EO7 microtubule-associated protein 4 1.42 H3139D12 cytoplasmic dynein intermediate chain 2 1.40 H3 076E07 unknown 1 .39 H3158GO6 erythroid differentiation regulator 1.37 H3085E11 unknown 1.33 H3130A07 unknown 1.32 H3132GO6 hypothetical 1 .28 H3068C02 unknown 1.27 H3032A01 translocase of inner mitochondrial membrane 1.26 H3085C07 zinc finger protein 1.26 H3117E10 ~ unknown 1.23 H3 150H1 1 hypothetical 1 .22 H3139C06 unknown 1.22 H3 047G07 unknown 1 .22 H3138CO7 unknown 1.21 H3041A07 no hit found 1.19 H3155A07 unknown 1.18 H3 046A02 unknown 1 .1 8 H3040C 12 unknown 1.15 H3001D07 unknown 1 . 12 TPlate identification number: sequence number for NIA 15K mouse cDNA library set 2The list of genes includes those that were significantly up-regulated by sulindac compared to control (P<0.05). Names written capital letters are for the respective homolog of the genes. The total number of genes that were up-regulated by sulindac is 53, which includes 19 co-regulated with anthocyanins (Table 13), 16 unknown, 6 no hit and 3 hypothetical protein genes. 124 3Ratio of intensity score; S/C: sulindac versus control. Ratio represents fold change in gene expression in tissue of mice consuming sulidac when compared that of mice consuming control. 125 Table 13. List of genes up-regulated by sulindac and anthocyanins in small intestine Plate ID1 Gene Name2 S/C3 A/C3 H3019H02 TAX INTERACTION PROTEIN 1 1.45 1.46 H3023H01 no hit found 1.36 1.38 H3155E09 hypothetical 1.36 1.39 H3102C12 ADP-ribosylation-like factor 6 interacting protein 1.30 1.22 H3125C12 guanine nucleotide binding protein, alpha inhibiting 3 1.29 1.20 H3145A05 hypothetical 1.27 1.29 mas proto-oncogene,insulin-like growth H3149AO9* factor type 2 receptor (Igf2r), L41ps, and Au76 peusogenes 1.26 1.13 H3149H02 unknown 1.26 1.17 H3066E10 no hit found 1.23 1.25 H306OE03 no hit found 1.23 1.29 H3077H06 LIM domain binding 1 1.21 1.79 H3026G06 tumor rejection antigen gp96 1.20 1.18 H3148H07 hypothetical 1.19 1.35 H3128B04 liver-specific bHLH-Zip transcription factor 1.19 1.22 H3108A01 interferon-stimulated protein 1.16 1.29 H3063GO7 unknown 1.16 1.40 H3039G11 unknown 1.13 1.13 H3109B07 unknown 1.13 1.10 H3155C06 heat stable antigen 1.12 1.22 TPlate identification number: sequence number for NIA 15K mouse cDNA library set 2The list of genes includes those that were significantly up-regulated by both sulindac and anthocyanins compared to control (P<0.05). Names written capital letters are for the respective homolog of the genes. The total number of genes that were up-regulated by both sulindac and anthocyanins is 19, which includes 4 unknown, 3 no hit and 3 hypothetical protein genes. 3 Ratio of intensity score; S/C: sulindac versus control, A/C: anthocyanins versus control. Ratio represents fold change in gene expression in tissue of mice consuming sulidac or anthocyanins when compared that of mice consuming control. *The true identity of the H3149A09 gene cannot be identified at this time and could be one of the four. 126 Table 14. List of genes down-regulated by sulindac vs control in small intestine Plate ID1 Gene Name2 C/S3 H3071D01 no hit found 1.68 H3086A12 unknown 1.54 H3154Bl 1 adipocyte-specific protein 5 1 .49 H3 146FO3 hypothetical 1 .46 H3123H07 novel cell death-regulatory protein GRIM19 1.44 H3107D06 unknown 1 .41 H3065F02 unknown 1 .40 H3033F 02 no hit found 1.38 H3134E01 hemoglobin, beta adult major chain 1.36 H3122H02 unknown 1 .36 H3122F10 putative THl protein 1.36 H3 142F09 hypothetical 1 .34 H3087F06 dynein 1.34 H3 108F07 alpha-L-iduronidase 1 .33 H3049B04 no hit found 1.32 H3136F06 unknown 1.32 H3132G02 hypothetical 1 .3 1 H3096H01 no hit found 1.30 H3138B03 unknown 1.30 H3156C08 metaxin 1.30 H3033D08 unknown 1.30 H3108D06 unknown 1 .29 H3 O2OF09 hypothetical 1 .29 H3045C02 ubiquitin-conjugating enzyme 1.28 H3 O60C 1 2 unknown 1 .28 H3025G06 hypothetical 1 .27 H3126H02 no hit found 1.27 H3045HO4 nonmuscle heavy chain myosin 1.27 H3060D01 CCR4-NOT transcription complex, subunit 7 1.27 H3065B01 hypothetical 1 .27 H3078G08 unknown 1.26 H3035D04 GM2 ganglioside activator protein 1.25 H3042BO6 unknown 1 .25 H3042F 12 serine (or cysteine) proteinase inhibitor 1.25 H3033A02 no hit found 1.25 H3108F08 programmed cell death 10 1.24 H3081FO3 hypothetical 1 .24 H3084B06 F -box protein 1.23 H3 O74C07 unknown 1.22 H3 140F02 unknown 1 .22 127 H3072C 12 unknown 1.21 H3015E11 PDZ and carboxyl terminal LIM domain 1 1.21 H3099H06 unknown 1 .20 H3 O44DO4 unknown 1 .20 H31 12BO6 unknown 1.20 H3020H08 unknown 1 .20 H3065B11 unknown 1.20 H3057D02 RhoGDI-l 1.19 H3016F02 unknown 1. 18 H3047G01 unknown 1.18 H3101F01 hypothetical 1.18 H3104D02 unknown 1 . 17 H3 06OF 1 2 hypothetical 1 .16 H3122A08 unknown 1 . 16 H3 096FO3 unknown 1 . 16 H3109F02 DREV protein 1.15 H3113F11 unknown 1.14 H3100G04 no hit found 1.14 H3 064BO6 no hit found 1 . l 3 H3077A12 hypothetical 1 . 1 1 H3104E04 no hit found 1 .07 TPlate identification number: sequence number for NIA 15K mouse cDNA library set 2The list of genes includes those that were significantly down-regulated by sulindac compared to control (P<0.05). The total number of genes that were down-regulated by sulindac is 108, which includes 47 genes co-regulated with anthocyanins (Table 15), 25 unknown, 9 no hit and 10 hypothetical protein genes. 3Ratio of intensity score; C/S: sulindac versus control. Ratio represents fold change in gene expression in tissue of mice consuming sulidac when compared that of mice consuming control. 128 Table 15. List of genes down-regulated by sulindac and anthocyanins in small intestine Plate 11)1 Gene Name2 as3 C/A3 H3104D07 fatty acid binding protein 3 2.34 1.78 H31 13812 no hit found 1.97 3.30 H3156A10unknown 1.84 2.16 2038 glutathione S transferase 1.67 1.69 H3007A01 unknown 1.66 1.56 H3091A01 hypothetical 1.65 1.39 H3105B07 hypothetical 1.52 1.38 H3124H12 no hit found 1.49 3.02 H3010H09 unknown 1.43 1.49 H3018G10unknown 1.42 1.39 H3004B03 no hit found 1.37 1.55 H3064F03 hypothetical 1.37 1.28 H3114G02 unknown 1.37 1.33 H3039E10 serine/threonine kinase 1.36 1.26 H3078F01 unknown 1.36 1.33 H3051E02 unknown 1.35 1.37 H3100F10 unknown 1.33 1.27 H3067F05 unknown 1.32 1.44 H3127G08 hypothetical 1.31 1.36 H3098E02 no hit found 1.30 1.26 H310OE10 hypothetical 1.30 1.36 H3143A07 unknown 1.28 1.46 H3097E12 unknown 1.28 1.20 H3084FO7 unknown 1.28 1.39 H3072E11 unknown 1.27 1.25 H3090D11 unknown 1.27 1.26 protein kinase, interferon inducible double H3024FO8 stranded RNA dependent activator 1.27 1.28 H3117A06 voltage-dependent calcium channel beta-3 subunit 1.26 1.17 H3142G12 hypothetical 1.26 1.13 H3057C12 hypothetical 1.26 1.35 H3088B01 hypothetical 1.26 1.26 H3008CO3 cytotoxic granule-associated RNA binding protein 1.25 1.28 H3047D07 nuclear receptor subfamily 5, group A, member 2 1.25 1.24 H3033D06 no hit found 1.24 1.31 H3142CO6 hypothetical 1.23 1.13 H309OFO7 unknown 1.23 1.14 H3100H05 unknown 1.22 1.30 H3119B07 unknown 1.21 1.23 129 H3126F07 unknown 1.21 1.19 H3052A08 unknown 1.19 1.26 H3065CO6 unknown 1.15 1.12 H3035G10unknown 1.15 1.19 H3132G08unknown 1.14 1.12 H3067B05 unknown 1.14 1.21 H3127G05 palmitoyl-protein thioesterase 1.13 1.17 H3032D10no hit found 1.12 1.11 H3134H05unknown 1.11 1.19 1Plate identification number: sequence number for NIA 15K mouse cDNA library set. Plate ID 2038 is not in the NIA 15K mouse cDNA library and added by Dr. John Lapres’ lab in Michigan State University. 2This list of genes includes those that were significantly down-regulated by both sulindac and anthocyanins compared to control (P<0.05). The total number of genes that were down-regulated by both sulindac and anthocyanins is 47, which includes 24 unknown, 6 no hit and 9 hypothetical protein genes. 3 Ratio of intensity score; C/S: sulindac versus control, C/A: anthocyanins versus control. Ratio represents fold change in gene expression in tissue of mice consuming sulidac or anthocyanins when compared that of mice consuming control. 130 DISCUSSION In this study, we confirmed our previous results in which we found that anthocyanins and cyanidin suppressed tumor development in cecum of ApcMin mice. In the previous experiments, the compounds (anthocyanins, cyanidin and sulindac) were administered in drinking water to a relatively small number (n=10) of mice per treatment group for 10 weeks. In this study, the extent to which dietary anthocyanins and sulindac inhibited intestinal tumor development was determined in a larger number of ApcMin mice (n=25) when fed compounds in the diet for 8 weeks. This approach increased the stability of the administered anthocyanins, increased power of the experiment by increasing the number of replicates, and reduced the morbidity of mice by shortening the promotion period. In this experiment, dietary anthocyanins significantly reduced adenoma number in all three intestinal sections. Cecal adenoma burden (volume) also was reduced and small intestinal adenoma mean diameter was intermediate in mice consuming anthocyanins. Sulindac reduced adenoma number and mean diameter in small intestine, but increased cecal adenoma number when compared to controls. Koide et a1. (1996) first reported that feeding red rice prolonged survival of Balb/C mice inoculated i.p. with Meth/A lymphoma cells and that hydrolyzed anthocyanins decreased the grth of HCT-15 cancer cells. Later, they found that sugar-bonded bioflavonoids extracted from red soybeans and red beans prolonged survival of Balb/C mice bearing tumor Meth/A cells and that anthocyanin fractions extracted from red soybeans inhibited the growth of HCT-l 5 cells (Koide et al., 1997). Pool-Zobol et a1. (1999) have shown that anthocyanins reduced H202- 131 induced DNA strand breaks but did not prevent endogenous generation of oxidized DNA bases in HT 29 clone 19A human colon cancer cells. Recently, it has been reported that purple corn anthocyanins suppressed the promotion of colon tumors caused by 2-amino—1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP) in rats initiated with dimethyl hydrazine. However, these researchers did not test the potential of purple corn color anthocyanins to directly suppress tumor development (Hagiwara et a1,2001) The second objective of this study was to determine the influence of dietary anthocyanins and sulindac on expression and cellular localization of B-catenin in the intestine of ApcMin mice. Contrary to our expectations, immunostaning of B-catenin in intestinal tissues demonstrated that sulindac generally did not influence the expression of B-catenin protein, whereas anthocyanins increased B-catenin expression in some intestinal regions compared to control or sulindac. Intracellular distribution of B-catenin was primarily in the membrane in all mice. These results are contradictory to the findings of other investigators. Mutanen et a1. (2000) M' - ‘" mice and decreased reported that rye bran diet prevented tumor formation in Ap‘c the cytosolic B-catenin level of intestinal epithelial cells. Cellular distribution of B- catenin also was modified by sphingolipid consumption (Schmelz et al., 2001). Sphingolipid feeding reduced adenoma numbers in all regions of the intestine in ApcMin mice and sphingosine reduced cytosolic and nuclear B—catenin expression (Schmelz et al, 2001). Gene expression results from the cDNA microarray analysis showed that expression of the mRNA transcripts for B-catenin and APC were not altered by 132 treatment, suggesting that transcriptional regulation did not cause the increase in B- catenin protein expression in the intestine of ApcMin mice consuming anthocyanins. Other possible explanations for the increase in B-catenin expression by anthocyanins would be increased stability of B-catenin mRNA, decreased degradation of B-catenin, orincreased translation. Additional experiments are necessary to address these possibilities. APC mutation is permissive for aberrant accumulation of cytoplasmic [3- catenin, which then translocates to the nucleus and binds ch/Lef transcription factors and causes increased transcriptional activation of genes in the Wnt/B-catenin signaling pathway. Known target genes of this signaling include PPARE, C-Myc, Cyclin D1 and MMP7. These genes have been found to be overexpressed in human colonic adenocarcinomas and in many human colonic tumor cell lines (He et al. 1999; Zhang et al., 1997; Crawford et al., 1999; Tetsu and McCormick, 2000; Newell et al., 1994). Our interest in testing the potential of tart cherry anthocyanins and cyanidin to inhibit tumor development in ApcMin mice stemmed from the observation that these compounds inhibit the activities of COX enzymes (Wang et al., 1999). McEntee et a1. (1999) reported that NSAID-induced regression of intestinal adenomas is associated with reduced levels of B-catenin. NSAIDs also have been shown to inhibit PPARS activity in HCT116 and SW480 cell lines in a dose- dependent manner (He et al., 1999). Therefore, we hypothesized that anthocyanins reduce tumor development in ApcMin mice in a manner similar to sulindac by suppressing Wnt/B-catenin signaling. 133 In contrast to our initial hypothesis, our results indicate that neither anthocyanins nor sulindac influenced the expression of mRNA of B-catenin and most of the predicted target genes of the Wnt/B-catenin signaling. Anthocyanin treatment modestly reduced the expression of cyclin D1 compared to sulindac in cecum but not in the other intestinal tissues. The expression of the other predicted target genes — MMP7, PPAR5 and C-Myc — were not altered by anthocyanins or sulindac in all intestinal sections of ApcMin mice. These results suggest that the mechanisms responsible for reductions in intestinal tumor development by sulindac or anthocyanins likely are not associated with inhibition of Wnt/B-catenin signaling. We consider it likely that tumor suppression associated with these compounds is via mechanisms other than normalized Wnt/B-catenin signaling. Although most hypotheses for explaining the anticarcinogenic effects of NSAIDs have focused on their common property to inhibit COX enzymes, the mechanism of the anticarcinogenic effect of these drugs has not been fully established. Little information on potential mechanisms whereby anthocyanins may suppress intestinal tumor development is available. Anthocyanins have been shown to inhibit the activity of epithermal growth factor receptor (EGFR) kinase. Meiers et al. (2001) demonstrated that two anthocyanins, cyanidin and delphinidin, are potent inhibitors of the EGFR kinase. Administration of inhibitors to the EGF R kinase also has been shown to reduce intestinal adenoma development in ApcMin mice (Torrance et al., 2000). A total of 1,492 genes were influenced by dietary treatment in any of the intestinal sections — small intestine, ceum or colon. A relatively small subset of 134 these gene effects overlapped across any two intestinal regions, and no single gene was influenced by treatment in all regions. To begin narrowing this list of gene changes to a shorter list of genes that merit future investigation, we have used two strategies. First, we identified genes influenced by treatment in more than one intestinal region. Second, we focused on gene changes that corresponded to the observed tumor numbers. For example, we are particularly interested in genes up- or down-regulated specifically by anthocyanins relative to sulindac and control treatments in the cecum or colon. Although gene changes are ranked in the tables by fold change, it is important to consider that an arbitrary selection of a fold change value as significant (i.e. 2-fold) is probably not appropriate for two reasons. First, all. gene changes reported in this experiment are based on statistical analysis of variance. Unlike many microarray experiments, in this study we have used true replication and valid statistical approaches to identify genes significantly influenced by treatment. Secondly, it is well established that the intestinal epithelium is a mixed cell population comprised by proliferating cells, terminally differentiated cells, and cells undergoing programmed cell death. Any changes in gene expression that occurred specifically in one of these cell types would be diluted by expression in the other epithelial cell types. We identified several clusters of differentially expressed genes in cecum, colon and small intestine of ApcMin mice using cDNA mcroarray analysis. Tumor inhibition by anthocyanins was most prominent in cecum. For this reason, we focused on genes changed specifically by anthocyanins relative to control and 135 sulindac in the cecum (Table 8). Although hemochromatosis gene and Ly6/Neurotoxin protein gene were influenced to the greatest extent by anthocyanins, there is very little information available on their functions or potential role in tumor development. Ras-related botulinum C3 substrate 1 also was up-regulated by anthocyanins. Clostridium botulinum C3 represents a major family of the bacterial toxins that transfer the ADP-ribose moiety of NAD to specific amino acids in acceptor proteins to modify key biological activities in eukaryotic cells, including protein synthesis, differentiation, transformation, and intracellular signaling (Han et al., 2001). C3-like transferases have been shown to ADP-ribosylate the small guanosine triphosphate (GTP)- binding protein family, Rho, resulting in functional inactivation of Rho (Wilde et al., 2001). Among the many genes altered by anthocyanins, we are particularly interested in those genes that potentially are involved in cellular differentiation (neuroendocrine differentiation factor), DNA synthesis and repair (ribonucleotide reductase M2), and regulation of apoptosis (translocase of inner mitochondrial ‘ membrane 8). Ribonucleotide reductases catalyze the production of the ' deoxyribonucleotides required for DNA replication and repair. Yamaguchi et al. (2001) demonstrated that a newly identified ribonucleotide reductase, p53R2, is directly regulated by p53 for supplying nucleotides to repair damaged DNA. Translocases of the inner mitochondrial membrane mediate transport of mitochondrial proteins from the cytosol into the mitochondria in conjunction with translocases of the outer mitochondrial membrane (Halestrap et al., 2000). Translocases of inner mitochondrial membrane also influence mitochondrial 136 membrane permeability, which is a critical step of apoptotic pathways (Crompton, 2000). Brenner et a1. (2000) demonstrated that adenine nucleotide translocator (ANT), the most abundant inner mitochondrial membrane protein, interacted with Bax, a proapoptotic member of the Bcl-2 family to yield an efficient composite channel. This opening of the mitochondrial permeability transition pore is thought to be involved in apoptosis. Altered intracellular signaling by increased expression of initiation factor 5, serine/threonine kinases (tetratricopeptide repeat domain, nemo-like kinase) and suppressor of mitogen-activated protein (MAP) kinase (astrocytic phophoprotein) also merit further investigation. Initiation factor 5 plays a role in translation initiation following recognition of the start codon (Das and Maitra, 2000). It has been shown that initiation factor 5 is required for hydrolysis of GTPase and acts as a GTPase-activator protein (Paulin et al., 2001). Nemo-like kinase is a serine- threonine protein kinase that connects MAP kinase and Wnt/B-catenin signaling pathways in Caenorhabditis elegans, Xenopus laevis and Drosophila melanogaster (Ishitani etal., 1999; Rocheleau et al, 1999; Verheyen et al., 2001). Ishitani et al. (1999) demonstrated that activation of TAKl-MAP kinase (a kinase activated by V transforming grth factor-beta) stimulated nemo-like kinase activity and downregulated Wnt signaling by inhibiting the interaction of the B-catenin-TCF complex with DNA. Astrocytic phosphoproteins are targets of numerous extracellular signals received by astrocytes. A major astrocytic phosphoprotein, PEA-15, has been identified as a protein kinase C substrate (Estelles et al., 1996). Formstecher et al. (2001) report that PEA-15 blocked epithelial receptor kinase 137 (ERK)-dependent transcription and proliferation by binding ERK5 and preventing their localization in the nucleus. Genetic deletion of PEA-15 also resulted in increased ERK nuclear localization with consequent increased cFos transcription and cell proliferation. No functional information is available on bladder cancer associated protein. Genes that were down-regulated in their expression by anthocyanins relative to control and sulindac include genes (Table 9) coding for G-protein coupled receptors (GTP-binding protein NGB, olfactory receptor cluster), cell adhesion proteins (nectin-3 alpha, beta-1,4-ga1actosyltransferase), enzymes involved in cell proliferation (omithine decarboxylase), an oncogenic protein (AS oncogene family), an apoptosis-related protein (Bcl-2-related ovarian killer protein), chromatin structural proteins (intermediate filament proteins family, chromatin structural protein), and a nucloesome assembly protein (spindlin). G-protein coupled receptors are cell surface receptors that are coupled to heterotrimeric G-proteins (GTP-bindin g proteins). They have binding sites in the extracellular domain for glycoprotein hormones. Down-regulated expression of these G-protein coupled receptors may indicate a reduction in intracellular signaling necessary for tumor growth. Beta-1,4- galactosyltranferase l is the key enzyme transferring galactose to the terminal N- acetylglucosamine (GlcNAc) of glycoconjugates (Furukawa and Sato, 1999). It also serves as a cell adhesion molecule by recognizing and binding to terminal GlcNAc of glycoconjugates on the adjacent cell surface and matrix. Reduced expression of omithine decarboxylase is also an important finding regarding the potential cellular actions of anthocyanins. Omithine decarboxylase is a rate-limiting enzyme for 138 synthesis of polyamines that ultimately regulate DNA synthesis (Williams-Adams et al., 1972). It is well known that intracellular polyamine concentrations and omithine decarboxylase activity are both increased in colorectal cancer tissue and in premalignant polyps (Wallace and Caslake, 2001). In colon, anthocyanin feeding reduced adenoma numbers compared to both control and sulindac treated mice. Anthocyanins also tended to reduce colonic adenoma volume compared to controls. Anthocyanins specifically increased the expression (Table 10) of genes coding for proteins involved in signal transduction (heat shock protein, transforming protein Rho A, interleukin 1 receptor-associated kinase, calcineurin B), transcription (GATA binding protein 2), mRN A splicing (transportin-SR), translation (458 pre rRNA), apoptosis control (BIMPl, inhibitor of growth family 1) and cell cycle control (cell cycle progression restoration 3). Heat shock proteins are considered to be molecular chaperones which play a universal role in maintaining cellular homeostasis (Liu and Steinacker, 2001). Rho genes encoding small GTP-binding proteins have been implicated in actin organization and the interaction of the cytoskeleton with intracellular membranes (Lackie and Dow, 1999). It has been reported that signaling and transforming properties of G protein coupled receptors cause aberrant grth of cells via activation of Rho family small GTPases (Whitehead etal., 2001). Interleukin 1 receptor-associated kinase (IRAK-l) is essential for pro-inflammatory cytokine interleukin l-beta signaling and interleukin 1 stimulation of nuclear factor KB (NF ~1cB), inducing an increase in the expression of many genes with roles in immunity and inflammation (Baldwin, 1996; Cooke et al., 2001). Calcineurin is a calmodulin-stimulated protein phosphatase and has a pivotal 139 role in the calcium/cahnodulin—calcineurin dependent pathway (Aramburu et al., 2000). Calcineurin, or PP2B, is a highly conserved, Ca2+ calmodulin-dependent phosphoserine/phosphothreonine-specific phosphatase and regulates many processes including NMDA signaling, Na+/K+ ATPase function, cardiac development and hypertrophy, T-cell activation, and angiogenesis (Cyert, 2001). GATA-binding proteins are a subfamily of zinc finger transcription factors with six members (GATA-L6) that interact with the GATA deoxyribonucleic acid (DNA) sequence. GATA sequences are found in the regulatory regions of many genes including those encoding T-helper 2-like cytokines, receptors, adhesion molecules and enzymes (Caramori et al., 2001). The GATA family of transcription factors also regulates tissue-specific patterns of gene expression during development by functioning as transcriptional effectors of the small GTPase RhoA (Charron et al., 2001). 45 S rRNA is structurally homologous to mammalian 7 S RNA and plays an essential role in targeting proteins containing a particular signal peptide to the secretory apparatus by forming signal recognition-like particle. It also binds independently to protein elongation factor G (EF-G), and functions in the translation process (N akamura et al., 2001). Transportin—SR proteins are serine/arginine-rich (SR) protein import receptors. SR proteins are a group of abundant arginine/serine (RS)—rich proteins and essential pre-mRNA splicing factors that are localized in the nucleus (Kataoka et al., 1999). The RS domain of these proteins serves as a nuclear localization signal and transportin-SR binds specifically and directly to the RS domains of several SR proteins (Lai et al., 2001). Inhibitor of growth family member 1(ING1) is a novel candidate tumor suppressor gene which is involved in the regulation of apoptosis (Ito 140 et al., 2002). INGl (p33) is thought to interact with the p53 signaling pathway and regulate cellular grth (Ito et al., 2002). McAllister-Lucas et al. (2001) characterized BHVIPI, a new signaling protein that binds BcllO and activates NF-KB. They found that BIMPl-mediated NF-kB activation required BcllO and IKB kinases. The involvement of BIMPl in the regulation of apoptosis is not clear. Interestingly, expression of S-adenosylmethionine decarboxylase (SAMDC) was increased by anthocyanins. SAMDC is involved in polyamine synthesis with omithine decarboxylase, implicating potential effects on cell proliferation in regulating colonic tumor grth (Milovica et al., 2001). The detailed functional information is not available for cell cycle progression restoration 3. Anthocyanins caused down-regulation (Table 11) of genes coding for proteins involved in signal transduction (inositol 1,4,5-triphosphate 3-kinase B, phospholysine phosphohistidine inorganic phyrophosphate phosphatase, A kinase anchor protein (yotiao) 9, orphan nuclear receptor (NR2E1), protein tyrosine phosphatase non-receptor type 21, ADP-ribosylation factor [GTP-binding proteins]), tumor suppression (deleted in colon cancer (DCC)), translational and post- translational modification (N-myristoyltransferase 1), apoptosis (neuronal apoptosis inhibitory protein 7), cell proliferation control (ribonuclease/angiogenin inhibitor 2) and a putative oncogene. Inositol 1,4,5-tn'phosphate 3-kinase mediates cellular signaling by phosphorylating the Cay-mobilizing second messenger inositol 1,4,5- triphosphate to yield the putative second messenger inositol 1,3,4,5-tetrakis phosphate (1P4). A kinase anchor protein (yotiao), a N-methyl-D-aspartate WMDA) receptor-associated protein, has been found to bind CAMP-dependent protein kinase 141 II (F eliciello et al., 1999). Yotiao has also been found to bind the type 1 protein phosphatase (PPl) and attaches PPl and CAMP-dependent protein kinase to NMDA receptors (Westphal et al., 1999). The downstream effect of the NMDA receptors related to yotiao protein is not clear. We found that anthocyanins decreased expression of the DCC gene, an unexpected observation since inactivation of DCC on chromosome 18 is known to be associated with colonic tumorigenesis and metastasis in humans (V ogelstein and Kinzler, 1993). Down-regulation of DCC by anthocyanins merits further study. N-myristoyltransferase catalyzes the co- translational and/or post-translational transfer of myristate to the amino terminal glycine residue of a number of important proteins. Maurer-Stroh et al. (2002) reported that N-myristoyltransferase catalyzes myristylation of kinases, phosphatases, proteasomal regulatory subunit 4, kinase interacting proteins and homologues of mitochondrial translocase. We focused on the gene changes caused by sulindac and, to a lesser extent, anthocyanins in small intestine because these compounds reduced small intestinal tumor development. Sulindac alone up-regulated genes for ATPase 6 subunit, nuclear protein (putative nuclear protein), DNA-binding proteins (kruppel-related zinc finger protein, zinc finger protein), microtubule-asscoiated proteins (microtubule-associated protein 4, cytoplasmic dyenin intermediate chain 2), and regulation of apoptosis (translocase of inner mitochondrial membrane) (Table 12). ATPase is an enzyme which yields ADP and inorganic phosphate by releasing the terminal phosphate from ATP. Sequence variants of the ATPase 6 gene are thought to play a role in mitochondrial DNA maintenance and integrity in yeast. Maximo et 142 al. (2001) reported that germline polymorphisms of the ATPase 6 gene are associated with mitochondrial DNA common deletion mutation, which increases thyroid tumorigenesis. Cytoplasmic dynein is a microtubule associated protein with ATPase activity involved in microtubule associated movement. The small intestinal genes up-regulated by both sulindac and anthocyanins (Table 13) include ones for adhesion (tax interaction protein 1), GTP—binding protein (ADP-ribosylation-like factor 6 interacting protein), GTP-binding inhibitor (GTP- binding protein alpha inhibiting 3), immune response (tumor rejection antigen gp96), and developmental decision (LIM domain binding). Tax protein has been identified as an axonal surface glycoprotein involved in cell adhesion. ADP-ribosylation factor is a ubiquitous GTP—binding protein. ADP-ribosylation is a form of post-translational modification of protein structure involving the transfer to protein of the ADP-ribosyl moiety of NAD (nicotinamide adenine dinucleotide). ADP-ribosylation is believed to play a part in normal cellular regulation. Tumor rejection antigen gp96 is a resident endoplasmic reticulum chaperone protein that directs peptides into the immune response pathway and assists in protein folding (Linderoth et al., 2000). Although the mechanism remains unclear, tumor rejection anti gen gp96 can activate the immune system to slow or stop the progression of tumors by escorting tumor- derived immunogenic peptides into the endogenous antigen presentation pathway of antigen presenting cells (Reed and Nicchitta, 2000). Among a total of 161 genes altered by sulindac in their small intestinal expression, there were more down-regulated genes (108) (Table 14 and 15) than up- regulated genes (53). Cell death regulation genes such as novel cell death-regulatory 143 protein GRIM19 and programmed cell death 10 were found to be down-regulated. Their exact function in cell death regulation has not been identified. Protein ubiquination and degradation may have been altered by down-regulation of ubiquitin-conjugating enzyme and F-box protein. Ubiquination has been identified as an important mechanism controling a diversity of cellular regulatory processes, including ubiquination-mediated protein turnover, protein targeting, and cell fate decisions. Clark et al. (1989) identified a developmentally essential gene, chB encoding ubiquiting conjugating enzyme, involved in substrate recognition in protein ubiquination. F-box protein is an adapter protein that is involved in associating proteins with the ubiquitin-driven proteolytic system. The F -box is a motif found in a wide variety of proteins including many cell cycle regulatory proteins, though various F-box proteins probably also play a part in regulation of transcription, signal transduction and development. Spiegelrnan et a1. (2002) reported regulation of F- box proteins by the Wnt/B-catenin signaling pathway. Activation of the Wnt/B- catenin signal transduction pathway elevates F-box protein BTrCP levels, whereas it reduces expression and activity of another closely related F -box protein, HOS. DNA helicase hFDH has been identified as an F-box protein (Kim et al., 2002). Down- regulation of DNA helicase hF DH influences regulation of DNA metabolism. Transcription and intracellular signal transduction are also influenced by altering chemokine receptor (CCR-NOT transcription complex), serine proteinase inhibitor, and PDZ and carboxyl terminal LIM domain 1. Genes that were down-regulated both by anthocyanins and sulindac (Table 15) included fatty acid biding protein (F ABP) 3 and glutathione S transferase. FABP 144 is thought play a role in enterocyte fatty acid trafficking by binding long chain fatty acids and facilitating exchange of fatty acids between intracellular membranes. Liver FABP is highly expressed in the both the liver and small intestine and its expression is likely up-regulated by PPAR5 activity in small intestine (Poirier et al., 2001). Poirier et al., (2001) demonstrated that the PPARS/PPAROL agonist GW2433 up-regulated the gene encoding liver FABP in the intestine of PPARa-null mice. Our results would indicate that anthocyanins and sulindac may alter the expression of genes that are downstream of PPARS signaling but not PPARB expression itself. Glutathione S transferase (GST) is a major phase II detoxification enzyme that catalyzes the conjugation of electrophilic substrates to glutathione. Although a protective role of the glutathione biotransforrnation in colorectal carcinogenesis has been suggested, studies that compared GST polymorphisms in relation to colon cancer risk are not conclusive with respect to an increased or decreased risk associated with a particular genotype (Strange and Fryer, 1999; Wormhoudt et al., 1999; Grubben et al., 2001). We were also interested in studying genes that were similarly influenced by treatment in different intestinal sections to enable us to identify patterns of gene changes that may be globally related to tumor development in the intestine. It was somewhat surprising that relatively few genes were similarly influenced by treatment in the different intestinal sections. However, the profiles of adenoma development and effects of treatment on tumor development were different throughout the intestine. No single gene was influenced by treatment in all three intestinal sections (small intestine, colon and cecum). The expression patterns of genes influenced by 145 treatment in more than one region of the intestine ofien were not consistent in the various regions. Our observations suggest that the mechanisms controlling tumor promotion in ApcMin mice likely differ in the various regions of the intestine. This hypothesis is supported by the distinct morphology of tumors in the small intestine versus the cecum and colon. In summary, we have demonstrated that dietary anthocyanins inhibit tumor Min development in small intestine, colon and cecum of Apc mice. Sulindac primarily inhibited small intestinal tumor development of ApcMm mice. Tumor inhibition by anthocyanins and sulindac does not appear to be associated with reductions in cytoplasmic B-catenin expression or reduced activity of the Wnt/B-catenin signaling pathway. cDNA microarray results showed that most of the predicted target genes of Wnt/B-catenin signaling were not influenced by anthocyanins or sulindac, suggesting the presence of downstream compensatory mechanisms for tumor inhibition by these compounds. We identified several genes that were differentially expressed in mice exposed to the three dietary treatments in intestinal sections. Genes influenced by anthocyanins and sulindac are involved in a wide variety of cellular and molecular processes such as DNA synthesis and repair, transcription, mRNA processing, protein interactions and turnover, cell proliferation and differentiation, cell cycle control, and programmed cell death (apoptosis). The patterns of gene expression changes suggest that alterations of cellular and molecular processes by anthocyanins and sulindac likely are different in ApcMin mice. Additional confirmatory analysis of these gene expression results will be necessary to further establish potential mechanisms whereby anthocyanins and sulindac influence tumor development. 146 CHAPTER VI OVERALL SUMMARY AND CONCLUSIONS 147 Anthocyanins, a member of the flavonoid family of phytochemicals, are widely distributed in plants including beans, fruits, and vegetables (Harbome and Grayer, 1988). We hypothesized that tart cherry anthocyanins would suppress intestinal tumorigenesis because they were previously found to inhibit the activities of COX enzymes (Wang et al., 1999b). Compounds that inhibit COX 2 activity have been demonstrated to protect against colon cancer development in animal models and reduce the risk of colon cancer in humans (Giovannucci et al., 1995; Labayle et al., 1991). However, little research has been conducted to study the potential of anthocyanins to inhibit colon cancer development. The first objective of this research was to determine the effects of tart cherry anthocyanins on the development of intestinal adenomas of ApcMin mice. ApcMin mice are a mutant mouse lineage predisposed to multiple intestinal neoplasia due to a mutation in the murine homolog of the APC gene (Su et al., 1992). The primary phenotype of ApcMin mice is the development of multiple adenomas throughout the small intestine, cecum and colon. Effective use of ApcMin mice in this research required an assessment of their normal process of tumor development. This was necessary to establish appropriate tumor endpoints for subsequent studies. There fore, we first examined the time of development and morphology of intestinal adenomas in ApcMin mice to obtain baseline data on tumor development. The first experiment was conducted to determine the influence of age and sex on the numbers and sizes of small intestinal, cecal and colonic tumors in ApcMm mice. Min Apc mice were weaned at 28 days of age and fed Teklad 8640 rodent diet 148 throughout the experiment. Mice were sacrificed at 30, 45, 60, 75, 90 and 120 days of age. As expected, the numbers and sizes of adenomas in the small intestine increased with age. The numbers of adenomas in the cecum and colon did not increase significantly with age, but the volume of colonic tumors increased significantly after 75 days of age. Sex did not influence the numbers of small intestinal, cecal or colonic tumors. Male mice had greater body weights, colon lengths and colon tumor volumes than female mice. Tumors were already present in 30 day-old‘mice. All small intestinal tumors were sessile (flat), whereas most cecal and colonic tumors were polypoid. These results provided important information that intestinal tumors develop early in life and progress rapidly in size as ApcMm mice exceed 90 days of age. We next examined the effects of tart cherry diets, anthocyanins, and their Min aglycone, cyanidin, on multiplicity and size of intestinal adenomas in Apc mice. Mi“ mice (n=10/treatment) were exposed During a 10 week experimental period, Apc to the following treatments; 1) control diet (modified AIN 93G diet containing 220 g/kg protein, 150 g/kg soybean oil and 50 g/kg cellulose), 2) control diet + 800 mg/L anthocyanins in the drinking water, 3) control diet + 200 mg/L cyanidin in the drinking water, 4) control diet + 200 mg/L sulindac in the drinking water, and 5) modified control diet containing 200 g/kg freeze-dried pitted tart cherries. ApcMin mice consuming anthocyanins, cyanidin, and tart cherries had significantly fewer numbers and smaller size of tumors in the cecum. Mice consuming sulindac had smaller small intestinal tumors, whereas mice consuming tart cherries had bigger 149 small intestinal tumors in average diameter. The total numbers and burden of colonic tumors were not significantly influenced by treatment. We also determined the potential of tart cherry anthocyanins, their aglycone, cyanidin, and their degradation products to directly inhibit growth of the human colon cancer cell lines HT 29 and HCT 116. Both anthocyanins and cyanidin reduced the growth of human colon cancer cells. The concentrations of anthocyanins and cyanidin required to cause a 50% inhibition of cell growth was 780 uM and 63 uM for HT 29, respectively and 285 uM and 85 11M for HCT 116, respectively. Anthocyanin degradation products did not influence cell growth. These results indicated that cyanidin directly inhibits the growth of colon cancer cells. Based on these results, we conducted a second feeding study with ApcM'" mice to determine the extent to which dietary anthocyanins and sulindac 1) inhibit intestinal tumorigenesis, 2) influence expression and cellular localization of B- catenin, and 3) influence expression of target genes of the Wnt/B-catenin signaling pathway as well as global gene expression profiles. The dietary treatments were; 1) control diet (modified American Institute of Nutrition 93G diet containing 220 g’k g protein, 150 g/kg soybean oil and 50 g/kg cellulose), 2) control diet + 200 ppm sulindac, 3) control diet + 750 ppm anthocyanins. The concentrations of sulindac (200 ppm) and anthocyanins (750 ppm) were based on efficacious concentrations used in the previous experiment, but were administered in the diet in this study to increase their stability. The results of this second study demonstrated that dietary anthocyanins significantly reduced adenoma development in all three intestinal sections — small 150 intestine, cecum and colon. Mice consuming anthocyanins had less cecal adenoma burden (volume) compared to mice consuming either sulindac or the control diet. Relative to controls, sulindac-consuming mice had fewer and smaller adenomas in small intestine, but larger cecal adenomas. Sulindac did not influence the expression of B-catenin protein in intestinal epithelium. Anthocyanins increased B-catenin levels in the middle small intestine and cecum compared to control or sulindac. Intracellular distribution of B-catenin was primarily in the cell membrane of mice consuming all treatments. cDNA microarray analysis showed that the expression of mRNA for APC, B-catenin, and most of the target genes of Wnt/B-catenin signaling were not influenced by treatment. Collectively, these observations indicate that the mechanisms responsible for tumor inhibition by sulindac in small intestine and by anthocyanins in all three intestinal sections do not directly involve inhibition of the Wnt/B-catenin signaling pathway. cDNA microarray analysis was used to screen for global profiles of gene expression that were influenced by anthocyanins and sulindac. A total of 1,492 genes were influenced by treatment in any of the intestinal regions. Relatively few genes were influenced by treatment in any two of the three intestinal sites — small intestine, cecum or colon. No single gene was influenced by treatment in all three regions. Genes influenced by anthocyanins and sulindac are involved in a wide variety of cellular and molecular processes such as DNA synthesis and repair, transcription, mRNA processing, protein interactions and turnover, cell proliferation and differentiation, cell cycle control, and programmed cell death. Among the many genes whose expression was influenced by treatment, we were particularly interested 151 in the following effects. Anthocyanins and sulindac up-regulated translocase of inner mitochondrial membrane 8 (regulation of apoptosis) in cecum and small intestine, respectively. Anthocyanins up-regulated ribonucleotide reductase M2 (DNA synthesis and repair) and nemo-like kinase, astrocytic phosphoprotein (signal transduction) whereas anthocyanins down-regulated omithine decarboxylase (cell proliferation). In summary, a series of experiments was conducted to determine the effects Min of anthocyanins on intestinal tumor development in Apc mice. Our most important findings were 1) characterization of the early and rapid development and Min progression of intestinal tumors in Apc mice, 2) demonstration that anthocyanins and cyanidin inhibited tumor development in ApcMin mice, 3) tumor inhibition by anthocyanins throughout the intestine of ApcMin mice, whereas sulindac only inhibited small intestinal tumors, 4) evidence that tumor inhibition in ApcMin mice by anthocyanins and sulindac is not likely to be mediated through inhibition of the Wnt/B-catenin growth signaling pathway, and 5) identification of several genes that may be related to reduced tumor development by anthocyanins and sulindac. Future experiments to further elucidate the effect of anthocyanins on intestinal development should include 1) confirmatory analysis using quantitative PCR or Northern blot analysis is needed to quantitate the changes in expression of genes identified by microarray analysis as being influenced by anthocyanins and/or sulindac, 2) cellular expression of signaling proteins and other markers associated with anthocyanin treatment by immunohistochemistry, 3) studies using other colon cancer models to confirm that anthocyanins inhibit colon cancer development, 4) 152 efficacy studies with compounds that are structurally related to anthocyanins but have greater stability. 153 APPENDICES 154 Table A1. Carrier status, body weights, and tissue lengths of mice from experiment I. Animal ID Sex Age Statusa Bwt (g) SI length (mm)b Colon length (mm) l M 30 C 14.0 303 64 2 M 30 C 14.9 317 75 3 F 30 C 12.9 305 79 4 F 30 C 16.8 309 71 5 M 30 C 14.9 316 79 6 F 30 C 10.0 289 59 7 F 30 C 14.3 290 55 8 F 30 C 15.0 288 66 9 M 30 N 16.5 288 66 10 F 30 N 15.3 329 64 11 M 30 N 16.3 320 68 12 F 30 N 16.3 318 70 13 M 30 N 19.7 350 75 14 F 30 N 14.8 314 63 15 F 30 N 14.0 309 49 16 F 30 N 15.0 350 72 17 M 30 N 10.2 265 53 18 M 30 N 10.5 252 64 19 M 30 N 11.6 285 64 20 M 30 N 17.5 334 82 21 M 30 N 14.4 311 76 22 M 30 N 13.9 304 76 23 M 45 C 19.0 342 83 24 F 45 C 18.2 334 80 25 M 45 C 16.7 320 74 26 M 45 C 20.8 320 81 27 M 45 C 18.2 300 80 28 F 45 C 15.8 295 71 29 M 45 C 19.4 333 83 30 F 45 C 16.9 310 73 31 M 45 C 19.0 325 79 32 M 45 C 18.5 335 67 33 M 45 N 20.5 310 72 34 M 45 N 20.5 342 81 35 F 45 N 17.0 302 74 36 M 45 N 21.5 309 85 37 M 45 N 21.0 330 85 38 F 45 N 16.0 325 70 39 M 45 N 18.9 339 74 40 M 60 C 19.5 303 69 155 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 6O 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 6O 60 60 6O 60 60 6O 60 6O 60 6O 6O 6O 60 6O 60 75 75 75 75 75 75 75 75 75 75 75 75 75 75 75 75 75 75 75 75 75 75 90 90 90 90 90 000002ZZZZZZZZZZZZOOOOOOOOOZZ22222200000000 24.3 21.3 17.5 18.9 20.7 18.3 18.8 21.1 21.9 23.9 19.7 22.6 19.5 18.4 18.9 23.5 23.1 19.1 21.2 21.5 19.4 20.7 21.9 18.5 19.0 24.5 20.2 24.5 19.0 24.3 24.8 20.3 20.6 18.3 19.7 21.2 17.8 21.3 25.4 19.2 20.5 21.1 23.2 338 333 324 346 340 322 345 335 333 380 309 325 370 340 349 365 362 357 330 356 330 370 330 339 347 340 355 370 327 374 366 360 350 353 343 356 325 336 360 330 345 362 318 85 94 ‘ 69 80 81 70 80 70 80 97 75 76 78 75 82 89 84 78 81 85 80 79 85 80 77 75 78 85 72 80 78 78 82 73 82 83 83 90 81 80 72 75 83 156 84 F 90 C 18.9 337 71 85 F 90 C 19.3 354 75 86 M 90 C 22.2 350 87 87 F 90 C 17.5 300 71 88 M 90 C 21.4 317 89 89 F 90 N 20.4 356 76 90 M 90 N 24.7 379 98 91 M 90 N 25.9 360 55 92 F 90 N 21.7 356 62 93 M 90 N 27.6 391 89 94 F 90 N 20.0 361 83 95 M 90 N 25.2 360 70 96 F 90 N 22.8 373 75 97 M 90 N 27.5 370 75 98 M 90 N 25.1 340 63 99 M 90 N 23.4 356 87 100 M 90 N 25.1 370 80 101 F 90 N 21.1 330 81 102 F 120 C 21.6 366 81 103 F 120 C 21.6 380 70 104 M 120 C 27.3 376 70 105 F 120 C 23.5 365 85 106 M 120 C 22.6 394 76 107 M 120 C 26.7 374 100 108 M 120 C 24.0 405 95 109 F 120 C 19.2 367 86 110 M 120 N 27.0 390 85 111 M 120 N 25.6 371 83 112 F 120 N 23.0 365 70 113 F 120 N 23.1 396 77 114 F 120 N 21.2 382 83 115 F 120 N 22.1 370 84 116 M 120 N 27.3 367 97 117 M 120 N 24.8 389 88 118 F 120 N 21.3 381 80 119 M 120 N 26.4 368 85 120 M 120 N 26.5 374 95 121 F 120 N 21.5 386 85 122 M 120 N 26.8 360 90 123 M 120 N 26.1 375 90 124 F 120 N 20.3 380 86 “Mm aCarrier Status: C; carrier (Apc bSmall intestine ), N; normal (Apc Table A2. Numbers and sizes of adenomas in ApcMin mice from experiment I. Adenoma Number Diameter Volumec (mm) (mm“) ID3 SIb PSIb MSIb DSIb Cecum Colon SIb Cecum Colon 1 6 3 0 3 2 O 5 3 O 2 8 3 3 2 0 l 8.5 0 l 3 13 6 3 4 0 3 10.5 0 1.5 4 10 3 4 3 l l 11 3 9 5 11 2 5 4 0 3 8 0 2.5 6 l3 4 5 4 2 2 13.5 4 2.8 7 16 5 5 6 0 1 18 0 0.8 8 42 7 l6 l9 0 O 25.5 0 0 23 25 2 10 13 1 3 21 1.5 2 24 27 6 11 10 l 2 20.5 1 5 25 25 6 12 7 1 2 22 2 3 26 35 7 12 16 1 0 29.5 1 0 27 26 3 13 10 l 3 21.5 1.5 2 28 34 9 13 12 1 O 28.5 1 O 29 31 3 16 12 2 2 24.5 2.5 3.5 30 46 11 21 14 3 2 42.5 3.5 l 31 65 10 26 29 0 1 49 0 1 32 46 2 27 17 2 2 31 2.5 2 40 10 4 2 4 O 2 13 O 1 41 9 3 5 1 O 1 11.8 0 0.5 42 24 3 5 16 4 1 24.5 4 0.5 43 25 4 7 14 O 1 21.5 0 l 44 25 4 8 13 O 1 28.8 0 0.5 45 27 3 10 14 0 0 29 0 0 46 34 5 14 15 3 1 33 4 0.5 47 108 9 21 78 2 2 80 1.5 5 48 101 14 34 53 2 4 91 2 4.5 57 29 4 10 15 l 1 29.5 1.5 l 58 25 6 l3 6 1 3 23 l 2.5 59 37 2 13 22 0 2 37 0 2 6O 50 8 15 27 1 l 48 2 1 61 43 5 16 22 2 2 40.5 3.5 l 62 78 5 22 51 2 2 67.5 3 2.5 63 58 7 23 28 2 4 61.5 4.5 3 64 53 9 25 19 0 2 50.5 0 5 65 90 38 38 14 l 3 96.5 1.5 2 79 10 4 2 4 0 2 13 0 1 8O 41 9 13 19 l 4 49 l 5 158 81 82 83 84 85 86 87 88 102 103 104 105 106 107 108 109 29 37 60 100 113 95 145 163 35 43 44 68 92 71 96 126 6 4 12 ll l6 17 19 11 "‘0th 0 7 14 15 16 30 32 40 45 62 7 ll 17 23 26 29 33 48 9 18 32 59 65 49 83 82 17 23 20 41 61 36 53 71 wH—‘wNNOWWNNH-bNo 0 MHw—‘OH—dONr—Av—sn—tmom 5 30 39.5 52 104.5 131 121 147.5 231 58.5 83 75.5 109 185.5 111 212.5 236 14 0 5.5 0 4 6 40 93.5 16.5 50.5 6 2.5 0 72 105 90 140 33.5 3 Animal ID. Note that Normal (Apcm) mice were excluded. b SI; small intestine, PSI; proximal small intestine, MSI; middle small intestine, DSI; distal small intestine cValues for adenoma volume of cecum and colon represent values calculated by width x length x height of adenomas. These values were transformed by 7t/ 6 to convert them to spherical tumor volumes in the final analysis. 159 Table A3. Weekly body weights (g) of ApcMin mice from the experiment 11. IDa Sex InitialkalWk2Wk3Wk4Wk5Wk6Wk7Wk8Wk9Wk10 Conl M 18.9 22.3 23.5 25.7 28.1 29.2 27.1 29.9 30.4 28.2 26.8 Con2 M 17.9 20.8 22.3 23.5 25.2 26.6 29.4 28.8 28.4 28.2 26.8 Con3 F 12.8 15.9 17.2 18.4 18 20 20.5 20 21.7 22.1 20.9 Con4 F 16.1 17.2 18.3 19.4 20.1 20.4 20.6 22.1 22.5 22.1 22.8 Con5 F 16.9 17.5 18.3 18.6 18.8 19.1 18.7 18.1 17.4 15.8 14.9 Con6 F 16.4 18.4 18.8 20.1 20.2 20.7 21.2 20.8 21.1 21.6 20.8 Con7 M 19.5 21.9 23.1 24.5 26.1 27 27.2 26.4 24.7 24.7 21.2 Con8 M 15.6 20.8 22.6 23.8 23.6 26.5 26 26.8 26.8 25.5 24.9 Con9 M 14.8 20.5 22.4 24.7 26.7 29.8 28.9 30.5 30.7 29.6 29.1 Antl M 19.5 23.5 24.9 25.6 24.2 29.5 31.1 32.2 33.7 35 36 Ant2 M 19.1 21.7 23.3 25.1 26.2 28.3 28.8 30.2 31.6 32.4 33.7 Ant3 F 16.2 18.1 18.9 19.6 20.2 20.6 20.4 17.8 16 NA NA Ant4 M 18.9 21.6 22.7 23.8 23.3 22.1 20.8 19.2 18.6 17.6 18.6 Ant5 F 15.5 17.2 19.2 20.4 20 21.4 21.4 21.8 19.8 17.1 15.3 Ant6 M 19.9 21.7 23.7 24.3 25.2 26.7 27.9 29.2 29.5 31.7 32.4 Ant7 F 15.6 17.2 19.1 19.4 20 21.2 21.8 22.5 22.6 22.7 22.9 Ant8 F 15.2 17.5 19.3 19.9 20.8 21.7 22 22 21 19.5 18.1 Cyal M 18 21.2 22.1 22.4 23.7 23.9 23.8 22.9 22.8 22.2 20.5 Cya2 F 17.2 19.5 20.3 21.5 23.3 22 23.2 27 26.5 23.8 26.2 Cya3 F 15.4 17.2 18.3 18.9 20.2 19.2 20.1 20.1 20.2 19.3 18 Cya4 M 18 20.7 21.8 22.6 22.5 23.1 22.5 21.6 18.9 18.9 19.5 Cya5 F 16.7 19 20.1 21.2 21.5 22.1 22.9 23.5 22.8 22.8 24 Cya6 F 16.6 16.8 19 20.4 20.5 21.1 22 22.4 22.6 22.5 22.5 Cya7 F 16.3 17.6 19.4 19.8 20.7 21.3 22.3 21.9 22.4 23 22.5 Sull M 16 19.8 21.3 22.3 22.8 23.5 25 25.5 25.9 26.4 27.1 Su12 F 16.2 17.6 19.1 20 21 20 21.1 22 23.1 22.6 22.8 Sul3 F 16.2 17.2 18.6 20.8 20.8 20.6 22.5 22.6 23.3 22.9 22.1 Sul4 M 16.5 19.9 21.4 21.8 22.4 23.4 23.6 24.4 25.4 22.1 25.3 Su15 F 15.7 17.1 18.3 19.5 20.7 21.2 21.9 21.7 22.1 25.3 22.3 Sul6 M 18 20.6 22.2 23.1 23.8 25 25.5 26.5 26 24.8 25.2 Sul7 F 17 17.8 19.4 19.7 20.7 21.4 21.3 21.6 22.1 22.2 22.4 Su18 F 15.3 17.5 19.3 19.7 19.8 20.2 21.5 23.4 23.9 25.8 26.7 Sul9 F 13.2 16.6 17.2 18.2 19.8 19.5 20.6 22.6 22.4 22.3 22.7 Sul 10 M 19 22.6 24.3 25.9 27.5 28.1 29 31.2 31.9 32.9 33.7 Chel M 17.5 20.3 22.2 23.2 23.3 24.2 24 26.1 26.4 26 24.5 Che2 F 13.9 17.9 19.9 19 20.7 21.5 22.2 23 22.3 22.9 23.5 Che3 F 15.9 17.5 18.6 19.9 19.9 17.3 16.7 15.2 15.7 15 NA Che4 M 19.3 21.3 22.2 22.5 23 22.4 21 17.7 19.7 14.3 NA CheS M 18.9 21.1 22.1 22.1 22.9 22.7 23.4 23.5 19.5 NA NA Che6 M 19.4 20.5 20.7 22.4 23 23.5 23.6 24.4 24.4 23.9 18.9 160 Che7 M 19.4 20.6 21.2 21.7 22.3 23 23.3 23.9 23.7 23.2 19.2 Che8 F 17.4 17.7 19.5 20.4 21.4 21.6 21 182 16.8 17.2 NA Che9 F 13.9 17.3 19.2 20.1 21.8 20.5 20.3 21.8 21.3 21 25.3 ChelOF 14 17.5 18.5 19.6 20.3 22 22.3 20.4 22.5 20.8 NA aAnimal ID: Con; control, Ant; anthocyanins, Cya; cyanidin, Sul; sulindac, Che; 20% Cherry diet. bInitial; body weight at the beginning of the experiment, week 10; body weight at the end of the experiment. NA: Data not available due to death of animal 161 Table A4. Tissue lengths and numbers and sizes of adenomas in ApcMm mice from experiment II. Lglgth Adenoma Number Diameter Volume (mm) (mm“) Z ;> Z > > ZZOOOZZOOOZZOOO HNHND—‘b—‘NHb—OI—i WN-kwwa-DLHNUJNZNNUJ OOOOOOb—‘OOOOZOOO Nt—ot—tt—INI—IWNF—‘NH 176 Table A9. Intensity scores of B-catenin expression in colon of mice from experiment IH. 11)“ Status“ Topc Middle“ Bottomc M C M C M C Con 40 C 2 1 1 O 1 0 Con 44 C 3 2 1 0 1 1 Con 49 C 3 2 2 1 2 1 Con 42 N NA NA NA NA NA NA Con 50 N 3 2 1 0 1 0 Sul 43 C 3 1 2 1 2 1 Sul 46 C 3 1 2 0 2 0 Sul 47 C 3 2 1 1 1 1 Sul 40 N 2 1 0 0 0 0 Sul 50 N 3 1 1 1 1 1 Ant 45 C 2 1 0 1 1 1 Ant 48 C 4 2 2 2 -2 1 Ant 49 C 4 3 1 1 2 1 Ant 40 N 2 1 1 0 1 0 Ant 50 N 3 1 2 1 2 1 aAnimal ID: Con; control, Ant; anthocyanins, Sul; sulindac bCarrier status: C; carrier (Apc+/M‘" ), N; normal (Apcm ) cTop; top one third of the crypts, Middle; middle one third of the crypts, Bottom; bottom one third of the crypts in the colon, M; membrane, C; cytoplasm 177 Table A10. Intensity scores of B-catenin expression in cecum of mice from experiment III. ID“ StatusB Topc Middlec Bottomc M C M C M C Con 40 C 3 1 1 O 1 0 Con 44 C 3 2 0 0 l 0 Con 49 C 4 2 1 0 0 0 Con 42 N 2 1 O O O 0 Con 50 N 3 2 2 1 O 0 Sul 43 C 3 1 2 O 1 0 Sul 46 C 2 1 0 O O O Sul 47 C 3 1 1 O 1 O Sul 40 N NA NA NA NA NA Na Sul 50 N 3 1 1 O 0 0 Ant 45 C 3 2 2 O 2 0 Ant 48 C 3 2 1 0 l 1 Ant 49 C 4 2 2 0 1 0 Ant 40 N 4 1 2 0 1 0 Ant 50 N 3 2 2 1 0 O alAnimal ID: Con; control, Ant; anthocyanins, Sul; sulindac bCarrier status: C; carrier (ApcWlin ), N; normal (Apcm ) °Top; top one third of the crypts, Middle; middle one third of the crypts, Bottom; bottom one third of the crypts in the cecum, M; membrane, C; cyt0plasm 178 LITERATURE CITED 179 American Cancer Society (2000) Cancer Facts and Figures-1997. ACS. Aramburu J, Rao A and Klee CB (2000) Calcineurin: from structure to function. Curr. Top. Cell Regul. 362237—95. Baker, SM, Harris, AC, Tsao, J -L, Falth, TJ, Bronner, E, Gordon, M, Shibata, D and Lisky, RM (1998) Enhanced intestinal adenomatous polyp formation in PmsZ"';Min mice. Cancer Res. 58:1087-1089. Baldwin, AS (1996) The NF-KB and IKB proteins: new discoveries and insights. Annu. Rev. Irnmunol. 14: 649-683. Barnes, CJ and Lee, M (1998) Chemoprevention of spontaneous intestinal adenomas in the adenomatous polyposis coli Min mouse model with aspirin. Gastroenterology 114: 873-877. Berenblum, I and Shubik, P (1947) The role of croton oil applications, associated with a single painting of a carcinogen, in tumor induction of the mouse skin. Br. J. Cancer 1: 379-382. Bilger, A, Shoemaker, AR, Gould, KA and Dove, WF (1996) Manipulation of the mouse germline in the study of Min-induced neoplasia. Sem. Cancer Biol. 7: 249- 260. Boolbol, SK, Dannenberg, A, Chadbum, C, Martucci, X, Guo, JT, Ramonetti, M, Abreu-Goris, HL, Newrnark, ML, Lipkin, DeCosse, JJ and Bertagnolli, MM (1996) Cyclooxygenase-2 overexpression and tumor formation are blocked by sulindac in a murine model of familial adenomatous polyposis. Cancer Res. 56:2556-2560. Brenner, C, Cadiou, H, Vieira, HL, Zamzabi, N, Marzo, I, Xie, Z, Leber, B, Andrews, D, Duclohier, H, Reed, JC and Kroemer, G (2000) Bcl-2 and Bax regulate the ~channel activity of the mitochondrial adenine nucleotide translocator. Oncogene 19: 329-336. Cademi, G., Bianchini, F, Dolara, P and Kriebel, D (1991) Starchy foods and colon proliferation in mice. Nutr. Cancer 15: 33-40. Cademi, G., Bianchini, F, Mancina, A, Spagnesi, MT and Dolara, P (1991) Effect of dietary carbohydrates on the growth of dysplastic crypt foci in the colon of rats treated with 1,2-dimethylhydrazine. Cancer Res. 51: 3721-3725. Caragay, AB. (1992) Cancer-preventive foods and ingredients. Food Tech. 46:65- 68. 180 Caramori, G, Lim, S, Ito, K, Tomita, K, Oates, T, J azrawi, E, Chung, KF, Bames, PJ and Adcock, IM (2001) Expression of GATA family of transcription factors in T- cells, monocytes and bronchial biopsies. Eur. Respir. J. 182466-73. Charron, F, Tsirniklis, G, Arcand, M, Robitailla, L, Liang, Q, Molkentin, JD, Meloche, S, Nemer, MT (2001) Tissue-specific GATA factors are transcriptional effectors of the small GTPase RhoA. Genes Dev. 15:2702-2719. Chiu, C-H, McEntee, MF and Whelan, J (1997) Sulindac caused rapid regression of preexisting tumors in Min/+ mice independent of prostaglandin biosynthesis. Cancer Res. 57: 4267-4273, Chomczynski, P and Sacchi, N (1987) Single-step method for RNA isolation by acid guanidium thiocyanate-phenol-chloroform extraction. Anal. Biochem. 162:156-159. Clark, A, Nomura, A, Mohanty, S and Firtel, RA (1997) A ubiquitin-conjugating enzyme is essential for developmental transitions in Dictyostelium. Mol. Biol. Cell 8: 1989—2002. Cooke, E.-L., Uings, LJ, Xia, CL, Woo, P and Ray, KP (2001) Functional analysis of the interleukin-1 B-stimulated nuclear factor KB (NF-KB) pathway activation: IPAK-l associates with the NF-KB essential modulator (NEMO) upon receptor stimulation. Biochem. J. 359: 403-410. Cormier, RT, Hong, KH, Halberg, RB, Hawkins, TL, Richardson, P, Mulherkar, R, Dove, WP and Lander, ES (1997) Secretory phospholipase Pla2g2a confers resistance to intestinal tumorigenesis. Nat. Genet. 17: 88-91. Crawford, HC, Fingleton, BM, Rudolph-Owen, LA, Heppner, KJ, Rubinfeld, B, Polakis, P and Matrisian, LM (1999) The metalloproteinase matrilysin is a target of B-catenin transactivation in intestinal tumors. Oncogene 18:2883-2891. Crompton, M (2000) Mitochondrial intermembrane junctional complexes and their role in cell death. J. Physiol. 529: 11-21. Cyert, MS (2001) Genetic analysis of calmodulin and its targets in Saccharomyces cerevisiae. Annu. Rev. Genet. 35:647-672. Das, S and Maitra, U (2000) Mutational analysis of mammalian translation initiation factor 5 (eIFS): role of interaction between the [3 subunit of eIF2 and eIFS in eIF function in vitro and in vivo. Mol. Cell Biol. 20: 3942-3950. Doll, R (1990) An overview of the epidemiological evidence linking diet and cancer. Proc. Nutr. Soc. 49: 119-131. 181 Duggan, DJ, Bittner, M, Chen, Y, Meltzer, P and Trent, JM (1999) Expression profiling using cDNA microarrys. Nature Gene Suppl. 21:10-14. Easwaran, V, Song, V, Polakis, P and Byers, S (1999) The ubiquitin-proteasome pathway and serine kinase activity modulate adenomatous polyposis coli protein- mediated regulation of beta-catenin-lymphocyte enhancer-binding factor signaling. 1 . Bio. Chem. 274: 16641-16645. Eklof—Spink, K, Fridman, SG and Weis, WI (2001) Molecular mechanisms of beta- catenin recognition by adenomatous polyposis coli revealed by the structure of an APC-beta-catenin complex. EMBO J. 20: 6203-6212. Elliot. (1992) Inhibition of glutathione reductase by flavonoids. A structure-activity study. Biochem. Pharrnacol. 8:1603-8. Estelles, A, Yokoyama, M, Nothias, F, Vincent, JD, Glowinski, J, Vernier, P, Chneiweiss, H (1996) The major astrocytic phosphoprotein PEA-15 is encoded by two mRNAs conserved on their full length in mouse and human. J. Biol. Chem. 271: 14800-14806. Fearon, ER and Vogelstein, B (1990) A genetic model for colorectal tumori genesis. Cell 61: 759-767. Feliciello, A, Cardone, L, Garbi, C, Ginsberg, MD, Varrone, S, Rubin, CS, Avvedimento, EV and Gottesman, ME (1999) Yotiao protein, a ligand for the NMDA receptor, binds and targets CAMP-dependent protein kinase 11(1). FEBS Lett. 464:174-178. Fodde, R, Edelmann, W, Yang, K, van Leeuwen, C, Carlson, C, Renault, B, Breukel, C, Alt, E, Lipkin, M, Khan, PM and Kucherlapati, R (1994) A targeted chain- termination mutation in the mouse Apc gene results in multiple intestinal tumors. Proc. Natl. Acad. Sci. USA 91: 8969-8973. Formstecher, E, Ramos, JW, Fauquet, M, Calderwood, DA, Hsieh, J C, Canton, B, Nguyen, XT, Bamier, JV, Carnonis, J, Ginsberg, MH, and Cheiweiss, H (2001) PEA-15 mediates cytoplasmic sequestration of ERK MAP kinase. Dev. Cell 1: 239- 250. Fujimori, M, Ikeda, S, Shimizu, Y, Okajima, M and Asahara, T (2001) Accumulation of B-catenin protein and mutations in exon 3 of B-catenin gene in gastrointestinal carcinoid tumor. Cancer Res. 61 :6656-6659. Furukawa, K and Sato, T (1999) Beta-1,4-ga1actosylation of N-glycans is a complex process. Biochem. Biophy. Acta. 1473: 54-66. 182 Giardiello, FM, Krush, AJ, Peterson, GM, Booker, SV, Kerr, M, Tong, LL and Hamilton, SR (1994) Phenotypic variability of familial adenomatous polyposis in 11 unrelated families with identical APC gene mutation. Gastroenterology 106: 1542- 1547.. Giovannucci, E, Egan, KM, Hunter, DJ, Starnpfer, MJ, Colditz, GA, Willett, WC and Speizer, FE (1995) Asprin and the risk of colorectal cancer in women. N. Engl. J. Med. 333: 609-614. Grubben, MJ, Nagengast, FM, Katan, MB and Peters, WH (2001) The glutathione biotransforrnation system and colorectal cancer risk in humans. Scand. J. Gastroenterol. Suppl. 234: 68-76. Hagiwara, A, Miyashita, K, Nakanishi, T, Sano, M, Tamano, S, Kadota, T, Koda, T, Nakamura, M, Irnaida, K, Ito, N and Shirai, T (2001) Pronounced inhibition by a natural anthocyanins, purple corn color, of 2-amino-1-methyl-6-phenylimidazo[4,5- b]pyridine (PhIP)—associated colorectal carcinogenesis in male F344 rats pretreated with 1,2-dimethylhydrazine. Cancer Lett. 171: 17-25. Halestrap, AP, Doran, E, Gillespie, JP and O’Toole, A (2000) Mitochondria and cell death. Biochem. Soc. Trans. 28: 170-177. Han, S, Arvai, AS, Clancy, SB and Tainer, J A (2001) Crystal structure and novel recognition motif of rho ADP-ribosylating C3 exoenzyme from Clostridium botulinum: structural insights for recognition specificity and catalysis. J. Mol. Biol. 305:95-107. Harbome, J. B. (1988) Flavonoids: Recent Advances. In: Plant Pigments, Goodwin, T. ed.; Academic Press; London, 299-343. Harbome, JB and Grayer, RJ (1988) The anthocyanins, in the flavonoids (Harbome, J B ed.) pp.1—20, Chapman and Hall, London. Hartwell, LH and Kastan, MB (1994) Cell cycle control and cancer. Science 266: 1821-1828. He, T-C, Chan, TA, Vogelstein, B and Kinzler, KW (1999) PPAR8 is an APC- regulated target of nonsteroidal anti-inflammatory drugs. Cell 99: 33 5-345. He, T-C, Sparks, AB, Rago, C, Herrneking, H, Zawel, L, Costa, LT, Morin, PJ, Vogelatein, B and Kinzler, KW (1998) Identification of c-MYC as a target of the APC pathway. Science 281: 1509-1512. 183 'I‘.""*n- . Hegde, P, Qi, Rong, Abernathy, K, Gay, C, Dharap, S, Gaspard, R, Earle-Hughes, J, Snesrud, E, Lee, N and Quackenbush, J (2000) A concise guide to cDNA microarray analysis 11. Biotechniques 29: 548-562. Hulsken, J, Birchmeier, W and Bchrens, J. (1994) E-cadherin and APC compete for the interaction with beta-catenin and the cytoskeleton. J. Cell. Biol. 127: 2061-2069. IARC (2001) GLOBOCAN 2000: Cancer Incidence, Mortality and Prevalence Worldwide, Version 1.0, CancerBase No. 5, Lyon, IARCPress. Ilyas, M and Tomlinson, [PM (1997) The interactions of APC, E-Cadherin and B- catenin in tumor development and progression. J. Pathol. 182: 128-137. Ishitani, T, Ninomiya—Tsuj i, J, Nagai, S, Nishita, M, Meneghini, M, Barker, N, Waterman, M, Bowerman, B, Clevers, H, Shibuya, H and Matsumoto K. (1999) The TAKl-NLK-MAPK-related pathway antagonizes signalling between beta-catenin and transcription factor TCF. Nature 399: 798-802. Ito, K, Kinjo, K, Nakazato, T, Ikeda, Y, Kizaki, M (2002) Expression and sequence analyses of p33(ING1) gene in myeloid leukemia. Am. J. Hematol 69: 141-143. J acoby, RF, Marshall, DJ, Newton, MA, Navarouic, K, Tutsch, K, Cole, CE, Lubet, RA, Kelloff, GJ, Verma, A, Moser, AR and Dove, WF (1996) Chemoprevention of spontaneous intestinal adenomas in the APC Min mouse model by the nonsteroidal anti-inflammatory drug proxicam. Cancer Res. 56: 710-714. Kataoka, N, Bachorik, J L and Dreyfuss, G (1999) Transportin-SR, a nuclear import receptor for SR proteins. J. Cell Biol. 145:1145-1152. Kennedy, BP, Payette, P, Mudgett, J, Vadas, P, Pruzanski, W, Kwan, M, Tang, C, Rancourt, DE and Cromlish, WA (1995) A natural disruption of the secretory group II phospholipase A2 gene in inbred mouse strains. J. Biol. Chem. 270:22378-22385. Kim, J, Kim, JH, Lee, SH, Kim, DH, Kang, HY, Bae, SH, Pan, ZQ and Seo, YS (2002) The novel human helicase hFDH is an F-box protein. J. Biol. Chem. In press. King, A and Young, G (1999) Characteristics and occurrence of phenolic phytochemicals. J. Am. Diet. Assoc. 99: 213-218. Kinzler, KW, Nilbert, MC, Vogelstein, B, Bryan, TM, Levy, DB, Smith, KJ, Preisinger, AC, Hamilton, SR, Hedge, P and Markham, A (1991) Identification of a gene located at chromosome 5q21 that is mutated in colorectal cancers. Science 251:1366-1370. 184 Kinzler, AR and Vogelstein, B (1996) Lessons from hereditary colorectal cancer. Cell 87:159-170. Kitagawa, M, Hatakeyama, S, Shirane, M, Matsumoto, M, Ishida, N, Hattori, K, Nakamichi, I, Kikuchi, A, Nakayarna, K and Nakayama, K (1999) An F-box protein, FWDl, mediates ubiquitin-dependent proteolysis of beta-catenin. EMBO J. 18: 2401-2410. Kennedy, AR, Beazer-Barclay, Y, Kinzler, KW and Newbeme, PM (1996) Suppression of carcinogenesis in the intestines of Min mice by soybean-derived Bowman-Birk inhibitor. Cancer Res. 56:679-682. Koide, T, Hashimoto, Y, Kamei, H, Kojima, T, Hasegawa, M, Terabe, K (1997) Antitumor effect of anthocyanins extracted from red soybeans and red beans in vitro and in vivo. Cancer Biother. Radiopharm. 12: 277-280. Koide, T, Kamei, Y, Hashimoto, T, Kojima, M, Hasegawa, M (1996) Antitumor effect of hydrolyzed anthocyanins from grape rinds and red rice. Cancer Biother. Radiopharm. 11: 173-177. Kristiansen, E, Meyer, 0 and Thorup, I (1996) Refined carbohydrate enhancement of aberrant crypt foci induced by the foodbome carcinogen 2-amino-3-methyl- imidazo[4,5-f]quinoline (IQ). Cancer Lett. 105: 147-151. Lai, MC, Lin, RI and Tarn, WY (2001) Transportin-SR2 mediates nuclear import of phosphorylated SR proteins. PNAS 98:10154-10159. Labayle, D, Fischer, D, Vielh, P, Drouhin, F, Pariente, A, Bories, C, Duhamel, O, Trousset, M and Attali, P (1991) Sulindac causes regression of rectal polyps in familial adenomatous polyposis. Gastroenterology 101: 635-639. Lackie, JM and Dow, JAT (1999) The dictionary of cell and molecular biology (third edition), Academic Press, London Lal, G, Ash, C, Hay, K, Redston, M, Kwong, E, Hancock, B, Mak, T, Kargman, S, Evans, JF and Gallinger, S (2001) Suppression of intestinal neoplasia mice by a specific cyclooxygenase 2 inhibitor and by a dual cyclooxygenase 1/2 inhibitor. Cancer Res. 61 :6131-6. Labarca, C and Paigen, K (1980) A simple, rapid, and sensitive DNA assay procedure. Anal. Biochem. 102: 344-352. Leister, I, Weith, A, Bruderlein, S, Cziepluch, C, Kangwangpong, D, Schlag, P and Schwab, M (1990) Human colorectal cancer: high frequency of deletions at chromosome 1p35. Cancer Res. 50: 7232-7235. 185 Leppert, M, Burt, R, Hughes, JP, Samowitz, W, Nakamura, Y, Woodward, S, Gardner, E, Lalouel, JM and White, R (1990) Genetic analysis of an inherited predisposition to colon cancer in a family with a variable number of adenomatous polyps. N. Engl. J. Med. 322: 904-908. Levi, S, Goodland, RA, Lee, CY, Stamp, G, Walport, MJ, Wright, NA, Hodgson, HJ (1990) Inhibitory effect of nonsteroidal anti-inflammatory drugs on mucosal cell proliferation associated with gastric ulcer healing. Lancet 336: 840-843. Levy, DB, Smith, KJ, Beazer-Barclay, Y, Harniltom, SR, Vogelstein, B and Kinzler, KW (1994) Inactivation of both APC alleles in human and mouse tumors. Cancer Res. 54: 5953-5958. Linderoth, NA, Popowicz, A and Sastry, S (2000) Identification of the peptide- binding site in the heat shock chaperone/tumor rejection antigen gp96(Grp94). J. Biochem. Chem. 25: 5472-5477. Lipken, M, Blattner, E, Fraumeni, JP, Lynch, HT, Deschner, E and Winawer, S (1983) Tritiated thymidine labeling distribution as a marker for the identification of hereditary predisposition to colon cancer. Cancer Res. 43: 1899-1904. Liu, CH, Chang, SH, Narko, K, Trifan, OC, Wu, MT, Smith, E, Haudenschild, C, Lane, TF and Hela, T (2001) Overexpression of cyclooxygenase 2 is sufficient to induce tumorigenesis in transgenic mice. J. Biol. Chem. 276: 18563-18569. Liu, Y and Steinacker, JM (2001) Changes in skeletal muscle heat Shock proteins: pathological significance. Front. Biosci. 6: D12-D25. Luongo, C, Moser, AR, Gledhill, S and Dove, WF ( 1994) Loss of Apc+ in intestinal adenomas from Min mice. Cancer Res. 54: 5947-5952. Martinsson, T, Weith, A, Cziepluch, C and Schwab, M (1989) Chromosome 1 deletions in human neuroblastoma: generation and fine mapping of microclones from the distal 1p region. Genes Chromosom. Cancer 1: 67-78. Mahmoud, NN, Carothers, AM, Grunberger, D, Bilinski, RT, Churchill, MR, Martucci, C, Newmark, HL and Bertagnolli, MM (2000) Plant phenolics decrease intestinal tumors in an animal model of familial adenomatous polyposis. Carcinogenesis 21: 921-927. Mas, T, Susperregui, J, Berke, B, Cheze, C, Moreau, S, Nuhrich, A and Vercauteren, J (2000) DNA triplex stabilization property of natural anthocyanins. Phytochemistry 53: 679-687. 186 Maurer-Stroh, S, Eisenhaber, B and Eisenhaber, F (2002) N-terrninal N- myristoylation of proteins: prediction of substrate proteins from amino acid sequence. J. Mol. Biol. 317:541-557. Maximo, V, Soares, P, Lima, J, Carneselle-Teijeiro, J and Sobrinho—Simoes, M (2002) Mitochondrial DNA somatic mutations (point mutations and large deletions) and mitochondrial DNA variants in human thyroid pathology: a study with emphasis on hurthle cell tumors. Am. J. Pathol. 160: 1857-1865. McAllister-Lucas, LM, Inohara, N, Lucas, PC, Ruland, J, Benito, A, Li, Q, Chen, S, Chen, FF, Yamaoka, S, Verma, 1M, Mak, TW and Nunez, G (2001) Bimpl , a MAGUK family member linking protein kinase C activation to BcllO-mediated NF- kappaB induction. J. Biol. Chem. 276:30589-30597. McCracken, JD, Wechter, WJ, Liu, Y, chase, RL, Kantoci, D, Murray, D, Quiggles, DD and Mineyama, Y (1996) Antiproliferative effects of the enantiomers of flurbiprofen. J. Clin. Pharrnacol. 36: 540-545. McEntee, MF, Chiu, C-H and Whelan, J (1999) Relationship of B-catenin and Bc1-2 expression to sulindac-induced regression of intestinal tumors in Min mice. Carcinogenesis 20: 635-640. McPhee, M, Chepenik, KP, Liddell, RA, Nelson, KK, Siracusa, LD and Buchberg, AM (1995) The secretory phospholipase A2 gene is a candidate for the Mom 1 locus, a major modifier of ApcM‘"- induced intestinal neoplasia. Cell 81: 95 7-966. Meiers, S (2001) The anthocyanidins cyanidin and delphinidin are potent inhibitors of the epidermal growth-factor receptor. J. Ag. Food Chem. 49: 95 8-62. Miller, EC and Miller, JA (1981) Mechanisms of chemical carcinogenesis. Cancer 47:1055-1064. Milovica, V, Turchanowa, L, Khomutov, AR, Khomutov, RM, Caspary, WF and Stein, J (2001) Hydroxylamine-containing inhibitors of polyamine biosynthesis and impairment of colon cancer cell growth. Biochem. Pharmacol. 61: 199-206. Miyazawa, T, Nakagawa, K, Kudo, M, Muraishi, K and Someya, K (1999) Direct intestinal absorption of red fruit anthocyanins, cyanin-3-glucoside and cyanin-3,5- diglucoside, into rats and humans. J. Agric. Food Chem. 47 :1083-1091. Mistry, TV, Cai, Y, Lilley, TH and Haslarn, E (1991) Polyphenol interactions, Part 5. Anthocyanin co-pigrnentation. J. Chem. Soc. Perkin. Trans. 2: 1287-1296. 187 Miyoushi, Y, Nagase, H, Ando, H, Horil, A, Ichii, S, Nakatsuru, S, Aoki, T, Miki, Y, Tori, T and Nakamura, Y (1992) Somatic mutations of the APC gene in colorectal tumorszmutation cluster region in the APC gene. Hum. Mol. Genet. 12229-233. Moolgavkar, SH and Knudson, AG (1981) Mutation and cancer: A model for human carcinogenesis. J. Natl. Cancer Inst. 66: 1037-1052. Morin PJ, Sparks AB, Korinek V, Barker N, Clevers H, Vogelstein B, Kinzler KW (1999) Activation of beta-catenin-ch signaling in colon cancer by mutations in beta- catenin or APC. 275: 1787-1790. Moser, AR, Pitot, HC and Dove, WF (1990) A dominant mutation that predispose to multiple intestinal neoplasia in the mouse. Science 247: 322-324. Munemitsu, S, Souza, B, Muller, O, Albert, 1, Rubinfeld, B and Polakis, P (1994) The APC gene product associates with microtubules in vivo and promotes their assembly in vitro. Cancer Res. 54: 3676-3681. Munemitsu, S, Albert, 1, Souza, B, Rubinfeld, B and polakis, P (1995) Regulation of intracellular B-catenin levels by the adenomatous polyposis coli (APC) tumor suppressor protein. Proc. Natl. Acad. Sci. USA 92: 3046-3050. Muscat, J E, Stellrnan, SD, Wynder, EL (1994) Nonsteroidal anti-inflammatory drugs and colorectal cancer. Cancer 74: 1847-1854. Mutanen, M, Pajari, A-M and Oikarinen, SI (2000) Beef induces and rye bran prevents the formation of intestinal polyps in ApcM‘" mice: relation to B-catenin and PKC isozyrnes. Carcinogenesis 21: 1167-1173. Nakamura, Y (1993) The role of the adenomatous polyposis coli (APC) gene in human cancers. Adv. Cancer. Res. 62:65-87. Nakamura, K, Miyamoto, H, Suzuma, S, Sakarnoto, T, Kawai, G, Yamane, K (2001) Minimal functional structure of Escherichia coli 4.5 S RNA required for binding to elongation factor G J. Biol. Chem. 276:22844-22849. Newell, KJ, Witty, JP, Rodgers, WH and Matrisian, LM (1994) Expression and localization of matrix-degrading metalloproteinases during colorectal tumori genesis. Mol. Carcinogenesis 10: 199-206. Nishida, T, Kaneko, F, Katagawa, M and Yasuda, H (2001) Characterization of a novel mammalian SUMO-1/Smt3-specific isopeptidase, a homologue of rat axam, which is an axin-binding protein promoting beta-catenin degradation. J. Biol. Chem. 276: 39060-39066. 188 h-m Ocke, MC, Bueno-de-Mesquita, I-IB, Feskens, EJ, van-Staveren, WA and Kromhout, D (1997) Repeated measurements of vegetables, fruits, beta-carotene, and vitamin C and E in relation to lung cancer. Am. J. Epidem. 145: 358-65. Olsson, NO, Caignard, A, Martin, MS, Martin, F (1984) Effect of indomethacin on the grth of colon cancer cells in syngeneic rats. Int. J. Irnmunopharrnacol. 6:329- 334. Oshima, M, Oshima, H, Kitagawa, K, Kobayashi, M, Itakura, C and Taketo, M (1995) Loss of APC heterozygosity and abnormal tissue building in nascent intestinal polyps in mice carrying a truncated Apc gene. PNAS 92: 4482-4486. Park, BH, Vogelstein, B and Kinzler, KW (2001) Genetic disruption of PPAR5 decreases the tumorigenicity of human colon cancer cells. PNAS 98:2598-2603. Paul, P, Letteboer, T, Gelbert, L, Groden, J, White, R and C0ppes, MJ (1993) Identical APC exon 15 mutations result in a variable phenotype in familial adenomatous polyposis. Hum. Mol. Genet. 2: 925-931. Paulin, FEM, Campbell, LE, O’Brien, K, Loughlin, J and Proud, CG (2001) Eukaryotic translation initiation factor 5(eIF5) acts as a classical GTPase-activator protein. Curr. Biol. 11:55-59. Paulsen, J E, Elvaas, I-KO, Steffenson, I-L an Alexander, JA (1997) Fish oil derived concentrate enriched in eicosapentanoic and docosahexanoic acid as ethyl ester suppresses the formation and growth of intestinal polyps in the Min mouse. Carcinogenesis 18: 1905-1910. Pierre, F, Perrin, P, Champ, M, Bomet, F, Meflah, K and Menanteau, J (1997) Short-chain fructo-oligosaccharides reduce the occurrence of colon tumors and develop gut associated lymphoid tissue in Min mice. Cancer Res. 57: 225-228. Pisani, P, Parkin, DM and Ferlay, J (1993) Estimates of the worldwide mortality from eighteen major cancers in 1985. Implications for prevention and projections of future burden. Int. J. Cancer 55:891-903. Pitot, HC (1989) Progression: The terminal stage in carcinogenesis. Jpn. J. Cancer Res. 80: 599-607. Pool-Zobel, BL, Bub, A, Schroder, N and Rechkemmer, G (1993) Anthocyanins are potent antioxidants in model systems but do not reduce endogenous oxidative DNA damage in human colon cells. Eur. J. Nutr. 38:227-34. 189 Potten, CS (1996) What is an apoptotic index measuring? A commentary. Br. J. Cancer 74: 1743-1748. Potten, CS (1997) Epithelial cell growth and differentiation. 11. Intestinal apoptosis. Am. J. Physiol. 273: G253-G257. Potter, JD, Slattery, ML, Bostick, RM and Gapstur, SM (1993) Colon cancer: A review of the epidemiology. Epidemiol. Rev. 15: 499-545. Prescott, SM and White, RL (1996) Self—promotion? Intimate connections between APC and prostaglandin H synthase-2. Cell 87:783-786. Ramirez-Tortosa, C, Anderson, OM, Gardner, PT, Morrice, PC, Wood, SG, Duthie, 51 SJ, Collins, AR, Duthie, GG (2001) Anthocyanin-rich extract decreases indices of lipid peroxidation and DNA damage in vitamin E-depleted rats. Free Radic. Biol. Med. 31:1033-1037. “" Reed, RC and Nicchitta, CV (2000) Chaperone-mediated cross-priming: a hitchhiker’s guide to vesicle transport (review). Int. J. Mol. Med. 6: 259-264. Reeves, PG., Nielsen, F H and Fahey, Jr., G.C (1993) AIN-93 purified diets for laboratory rodents: final report of the American Institute of Nutrition ad hoc writing committee on the reformulation of the AIN-76A rodent diet. J. Nutr. 123: 1939- 195 1 . Renner, HW (1990) In vivo eflects of single or combined dietary antimutagens on mutagen-induced chromosomal aberrations. Mutat. Res. 244: 185-188. Rice-Evans, CA, Miller, NJ, Bolwell, PG, Brarnley, PM and Pridham, JB (1995) The relative antioxidant activities of plant-derived polyphenolic flavonoids. Free Radical Res. 22:3785-93. Rigau, J, Pique, J, Rubio, E, Planas, R, Tarrech, JM, Bordas, JM (1991) Effects of long-terrn sulindac therapy on colonic polyposis. Ann. Intern. Med. 115: 952-955. Ritland, SR and Gendler, SJ (1999) Chemoprevention of intestinal adenomas in the ApcMin mouse by piroxicam: kinetics, strain effects and resistance to Chemoprevention. Carcinogenesis 20:51-58. Rocheleau, CE, Yasuda, J, Shin, TH, Lin, R, Sawa, H, Okano, H, Priess, JR, Davis, RJ and Mello, CC (1999) WRM-l activates the LIT-1 protein kinase to transduce anterior/posterior polarity signals in C. elegans. Cell 97: 717-726. Rosin-Arbesfeld, R, Townsley, F and Bienz, M (2000) The APC tumor suppressor has a nuclear export function. Nature 406:1009-1012. 190 Roy, HK, Karoski, WJ, Ratashak, A and Smyrl, TC (2001) Chemoprevention of intestinal tumorigenesis by nabumetone: induction of apoptosis and Bel-2 downregulation. Br. J. Cancer 18:1412-1416. Rubinfeld B, Souza, B, Albert, 1, Muller, O, Chamberlain, SH, Masiarz, FR, Munemitsu, S, Polakis P (1993) Association of the APC gene product with beta- catenin. Science 10:1731-1734. Rubinfeld, B, Albert, 1, Porfiri, E, Fiol, C, Munemitsu, S and Polakis, P (1996) Binding of GSK 313 to the APC-B-catenin complex and regulation of complex assembly. Science 272: 1023-1026. Salic, A, Lee, E, Mayer, L, Kirschner, MW (2000) Control of beta-catenin stability: reconstitution of the cytoplasmic steps of the wnt pathway in Xenopus egg extracts. Mol. Cell 5: 523-532. Sarrna, AD and Sharma, R (1999) Anthocyanin-DNA copigrnentation complex: mutual protection against oxidative damage. Phytochemistry 52: 1313-1318. Sasai, H, Masaki, M and Wakitani, K (2000) Suppression of polypo genesis in a new mouse strain with a truncated ApcA474 by a novel COX-2 inhibitor, J TE-522. Carcinogenesis 21:953-958. Seeram, N. P., Momin, R. A., Bourquin, L. D. and Nair, M. G. (2001) Cyclooxygenase inhibitory and antioxidant cyanidin glycosides from cherries and berries. Phytomedicine, 8: 362-369. Seeram, NP, Bourquin, LD and Nair, MG. (2001) Degradation products of cyanidin glycosides from tart cherries and their bioactivities. J. Agric. Food Chem. 4924924- 4929. Schmelz, EM, Roberts, PC, Kustin, EM, Lemonnier, LA, Sullards, C, Dillehay, DL and Merill Jr, AH (2001) Modulation of intracellular B-catenin localization and intestinal tumorigenesis in vivo and in vitro by sphingolipids. Cancer Res. 61 :6723- 6729. Sheng, H, Shao, J, Williams, CS, Pereira, MA, Taketo, MM, Oshima, M, Reynolds, AB, Washington, MK, DuBois, RN and Beauchamp, RD (1998) Nuclear translocation of B-catenin in hereditary and carcinogen-induced intestinal adenomas. Carcinogenesis 19: 543-549. Shoemaker, AR, Moser, AR and Dove, WF (1995) N-Ethyl-N-nitosourea treatment of multiple intestinal neoplasia (Min) mice: Age-related effects on the formation of intestinal adenomas, cystic crypts, and epiderrnoid cycts. Cancer Res. 55:4479-4485. 191 Shiff, SJ, Qiao, L, Tsai, LL and Rigas, B (1995) Sulindac sulfide, an aspirin-like compound, inhibits proliferation, causes cell cycle quiescence, and induces apoptosis in HT-29 colon adenocarcinoma cells. J. Clin. Invest. 96:491-503. Skinner, SA, Penney, AG, O’Brien, PE (1991) Sulindac inhibits the rate of growth and appearance of colon tumors in the rat. Arch. Surg. 126: 1094-1096. Smith, KJ, Levy, DB, Maupin, P, Pollard TD, Vogelstein, B and Kinzler, KW (1994) Wild-type but not mutant APC associates with the microtubule cytoskeleton. Cancer Res. 54:3672-3675. Sorenson, IL, Kristiansen, E, Mortensen, A, Nicolaisen, GM, Wijnands, J AH, van Kranen, H], van Kreijl, CF (1998) The effect of soy isoflavones on the development of intestinal neoplasia in ApcMin mouse. Cancer Lett. 130: 217-225. Speigelman, VS, Tang, W, Katoh, M, Slaga, TJ and Fuchs, SY (2002) Inhibition of HOS expression and activities by Wnt pathway. Oncogene 21: 856-860. Steffensen, I-L, Paulsen, JE, Eide, TJ and Alexander, J (1997) 2-amino-1-methyl-6- phenylimidazo[4,5-b]pyridine increase the numbers of tumors, cystic crypts and aberrant crypt foci in multiple intestinal neoplsia mice. Carcinogenesis 18:1049- 1054. Strange, RC and Fryer, AA (1999) Chapter 19. The glutathione S-transferase: influence of polymorphism on cancer susceptibility. IARC Sci. Pub]. 148: 231-249. Su, LK, Kinzler, KW, Vogelstein, B, Preisinger, AC, Moser, AR, Luongo, C, Gould, KA and Dove, WF (1992) Multiple intestinal neoplasia caused by a mutation in the murine homolog of the APC gene. Science 256: 668-670. Su, LK, Johnson KA, Smith, KJ, Hill, DE, Vogelstein, B and Kinzler, KW (1993a) Association between wild type and mutant APC gene products. Cancer Res. 53: 2728-2731 . Su, LK, Vogelstein, B and Kinzler, KW (1993b) Association of the APC tumor suppressor protein with catenins. Science 262: 1734-1737. Sugauma, M, Ohkura, Y, Okabe, S and Fujiki, H (2001) Combination cancer chemoprevention with green tea and sulindac shown in intestinal tumor formation in Min mice. J. Cancer Res. Clin. Oncol. 127:69-72. Takeichi, M (1988) The cadherins: cell-cell adhesion molecules controlling animal morphogenesis. Development 102:639-655. 192 Tanaka, TS, J aradat, SA, Lim, MK, Kargul, GJ, Wang, X, Grahovac, MJ, Pantano, S, Sano, Y, Piao, Y, Nagaraja, R, Doi, H, Wood, WH, Becker, KG and Ko, MS (2000) Genome—wide expression profiling of mid-gestation placenta and embryo using a 15,000 mouse developmental cDNA microarray. PNAS 97: 9127-9132. Taya, S, Yamamoto, T, Kanai-Azuma, M, Wood, SA and Kaibuchi, K (1999) The deubiquitinating enzyme Farn interacts with and stabilizes beta—catenin. Genes. Cells. 12: 757-767. Tetsu, O and McCormick, F (1999) B-catenin regulates expression of cyclin D 1 in colon carcinoma cells. Nature 398:422-426. Terpstra TO, van Blankenstein, M, Dees J, Eilers, GAM (1987) Abnormal pattern of cell proliferation in the entire colonic mucosa of patients with colon adenoma or cancer. Gastroenterology 92: 704—708. Torrance, CJ, Jackson, PE, Montgomery, E, Kinzler, KW, Vogelstein, B, Wissner, A, Nunes, M, Frost, P and Discafani, CM (2000) Combinatorial chemoprevention of intestinal neoplasia. Nat. Med. 6:1024-1028. Tsuda, T, Horio, F and Osawa, T (1999) Absorption and metabolism of cyanidin 3- O-B-D-glucoside in rats. FEBS Lett. 449: 179-182. Tsuda, T, Ohshima, K, Kawakishi, S and Osawa, T (1996) Oxidation products of cyanidin 3-O-B-D-glucoside with a free radical initiator. Lipids 31: 1259-1263. Tsuda T. (2000) The role of anthocyanins as an antioxidant under oxidative stress in rats. Biofactors 13:133-9. Tsuda T. (1998) Dietary cyanidin 3-O-beta-D-glucoside increases ex vivo oxidative resistance of serum in rats. Lipids 33:583-8. Van Kranen, HJ, van Iersel, PWC, Rijnkels, JM, Beems, DB, Alink, GM, van Kreijl, CF (1998) Effects of dietary fat and a vegetable-fruit mixture on the development of intestinal ne0plasia in the APC“ mouse. Carcinogenesis 19: 1597-1601. Verheyen, EM, Mirkovic, I, MacLean, SJ, Langrnann, C, Andrews, BC and MacKinnon, C (2001) The tissue polarity gene nemo carries out multiple roles in patterning during Drosophila development. Mech. Dev. 101: 119-132. Vogelstein, B and Kinzler, KW (1993) The multistep nature of cancer. Trends Genet. 9: 138-139. Wallace, HM and Claslake, R (2001) Polyarnines and colon cancer. Eur. J. Gastroenterol. Hepatol. 13: 1033-1039. 193 Wang, H, Nair, MG, Iezzoni, AF, Strasburg, GM, Booren, AM and Gray J I (1997) Quantification and characterization of anthocyanins in Balaton tart cherries. J. Agric. Food Chem. 45: 2556-2560. Wang, H, Nair, MG, Strasburg, GM, Booren, AM, and Gray, J I (1999a) Novel antioxidant compounds fi'om tart cherries (Prunus cerasus). J Nat. Prod. 62: 86-88. Wang, H, Nair, MG, Strasburg, GM, Chang, Y.-C. Booren, AM. Gray, J I and DeWitt, DL (1999b) Antioxidant and anti-inflammatory activities of anthocyanins and their aglycone, cyanidin, from tart cherries. J. Nat. Prod. 62: 294-296. Weiser, MM (1973) Intestinal epithelial cell surface membrane glycoprotein synthesis. 1. An indicator of cellular differentiation. J. Biol. Chem. 248:2536-2541. Westphal, RS, Tavalin, SJ, Lin, JW, Alto, NM, Fraser, ID, Langeberg, LK, Sheng, M and Scott, JD (1999) Regulation of NMDA receptors by an associated phosphatase- kinase signaling complex. Science 285: 93-96. WCRF/AICR (1998) Food, Nutrition and the prevention of cancer: A global perspective. Wilde, C, Chhatwal, GS, Schmalzing, G, Arktories, K and Just, I (2001) A novel C3-like ADP-ribosyltransferase from Staphylococcus aureus modifying RhoE and Rnd3. J. Biol. Chem. 276:9537-9542. Willet, WC (2001) Diet and cancer: one view at the start of the millennium. Cancer Epidemiol. Biomarkers Prev. 10: 3-8. Williarns-Adams, HG, Jana, J, Coppocc, GL, Geroch, ME and Sherone, AC (1972) New aspects of polyamine biosynthesis in eukaryotic organisms. Advances in Enzyme Regulation 10: 225-245. Whitehead, IP, Zohn, IE and Der, CJ (2001) Rho GTPase-dependent transformation by G protein-coupled receptors. Oncogene 20: 1547-1555. Worrnhoudt, LW, Commandeur, JN and Vermeulen, NP (1999) Genetic polymorphisms of human N-acetyltransferase, cytochrome P450, glutathione-S- transferase, and epoxide hydrolase enzymes: relevance to xenobiotic metabolism and toxicity. Crit. Rev. Toxicol. 29: 59-124. Wright, NA and Irwin, M (1982) The kinetics of villus cell populations in the mouse small intestine: normal villi-the steady state requirement. Cell Tissue Kinet. 15:595- 609. 194 Yamaguchi, T, Matsuda, K, Sagiya, Y, Iwadate, M, Fujino, MA, Nakamura, Y and Arakawa, H (2001) p53R2-dependent pathway for DNA synthesis in a p53- regulated dell cycle checkpoint. Cancer Res. 61 :8256-8262. Zhang, T, Nanney, LB, Luongo, C, Lamps, L, Heppner, KJ, DuBois, RN and Beauchamp, RD (1997) Concurrent overexpression of cyclin D1 and cyclin- dependent kinase 4(cdk 4) in intestinal adenomas from multiple intestinal neoplasia (Min) mice and human familial adenomatous polyposis patients. Cancer Res. 57: 169-175. 195 IIIIIIIIIIIIIIIIIIIIIIIIIIIIIII 1.11111lllljlllllllljllllljljlll