IIHIHLH ‘Ul WI .‘401‘ Ioooo ‘ _m4>oo meme ”tame lll‘lllllllll‘lllll‘ Un lversity This is to certify that the thesis entitled SPHINGOLIPIDS IN FOODS AND DIFFERENTIAL SENSITIVITY OF HUMAN COLON CANCER CELLS TO SPHINGOID BASES AND CERAMIDES presented by Eun-Hyun Ahn has been accepted towards fulfillment of the requirements for M.S. degree in Human Nutrition 7221283535?” Joseph J. Schroeder Date December 10, 1998 0-7639 MS U is an Affirmative Action/Equal Opportunity Institution r PLACE IN RETURN Box to remove this checkout from your record. To AVOID FINE return on or before date due. MAY BE RECALLED with earlier due date if requested. DATE DUE DATE DUE DATE DUE use W14 SPHINGOLIPIDS IN FOODS AND DIFFERENTIAL SENSITIVITY OF HUMAN COLON CANCER CELLS TO SPHINGOID BASES AND CERAMIDES by Eun-Hyun Ahn A THESIS Submitted to Michigan State University in partial fulfillment of the requirements for the degree of MASTER OF SCIENCE Department of Food Science and Human Nutrition 1998 ABSTRACT SPHINGOLIPIDS IN FOODS AND DIFFERENTIAL SENSITIVITY OF HUMAN COLON CANCER CELLS TO SPI;I)NGOID BASES AND CERAMIDES Eun-Hyun Ahn Complex sphingolipids have been shown to protect against development of colon cancer. Sphingoid bases and ceramides are bioactive metabolites of sphingomyelin and other more complex sphingolipids found in foods. The purpose of the present study was to quantitate sphingolipids in some common foods and investigate the effects of Sphingoid bases and ceramides on growth and death of HT-29 and HGT-116 human colon cancer cells. The concentrations of total sphingolipids and free Sphingoid bases (nmng of dry weight) were approximately: nonfat dry milk (203, 146); yogurt (138, 1.2); Swiss cheese (167, 6.5); full fat soy flakes (609, 2.6); soy flour (610, 1.6); isolated soy protein (210, 2.8). Most sphingolipids in foods were present as complex sphingolipids and sphingosine was the predominant Sphingoid base backbone of total sphingolipids in foods. Sphingosine, sphinganine, and Cz-ceramide inhibited grth and caused death of HT-29 and HCT-l 16 cells in concentration- and time-dependent manners; whereas, Cz-dihydroceramide had no effect suggesting that the 4,5-trans double bond was necessary for the inhibitory effect of ceramide. Sphingosine and sphinganine killed cells by inducing apoptosis; whereas, Cz-ceramide did not induce apoptosis. The results indicate that sphingolipids are significant constituents of soy fractions and dairy products and that the colonic concentrations of Sphingoid bases and/or ceramide which may be achieved after consumption of the foods are sufficient to inhibit growth and cause death of human colon cancer cells. ACKNOWLEDGEMENTS I would like to express very special thanks to my major advisor, Dr. Joseph J. Schroeder for his kind support, endless understanding, warm encouragement and wise guidance. I was lucky enough to be his first graduate student. I also would like to extend my sincere gratitude to my committee, Dr. Dale R. Romsos and Dr. Leslie D. Bourquin for their guidance. I would like to thank my fellow graduate students-Chi Zhang, Hong Yang, and Min-Sun Kim and Dr. Do-Yle Lee, Julie Arnold, Stacey Tremp, and Jason Wiesinger for their companionship. All of them contributed to various parts of my pleasant memories while we worked at the same laboratory. I cannot forget to give my appreciation to Dr. Won 0. Song for her loving encouragement. My thanks also go to Dr. James J. Pestka and Dr. John E. Linz who allowed me to use their instruments. Finally, I would like to give very sincere thanks to my precious family for their unlimited love, measureless understanding, patience, and support. Their presence have always brought me happiness and strength. I also thank my special friends in Seoul, Korea and USA for their friendship and encouragement. I dedicate this fruit of my sweat to my parent who gave me a life, my older brothers, relatives, special friends, and God who created the world. iii TABLE OF CONTENTS Page LIST OF TABLES v LIST OF FIGURES vi I. LITERATURE REVIEW 1 A. Colon cancer 1 l. Colon cancer incidence 2. Colon cancer etiology 3. Colon cancer and diet B. Dietary sphingolipids might reduce colon carcinogenesis 5 1. Sphingolipids regulate cell behavior 2. Milk sphingomyelin might prevent the progression of adenomas to adenocarcinomas in colon cancer 3. Ceramide and sphingosine may mediate the protective effect of sphingomyelin against colon cancer C. Knowledge of the types and concentrations of sphingolipids 14 in foods is limited II. OBJECTIVES 17 III. MATERIALS AND METHODS 18 IV. RESULTS 28 A. Sphingolipid concentrations in foods 28 B. Differential sensitivity of human colon cancer cells to 33 Sphingoid bases and ceramides V. DISCUSSION 41 VI. REFERENCES 47 iv Table 1. Table 2. Table 3. LIST OF TABLES Total lipids and moisture content in foods. Sphingolipid concentrations in foods. Percentage of free Sphingoid bases and Sphingoid base backbones of food complex sphingolipids that are sphingosine and sphinganine. Page 29 3O 32 Figure 1. Figure 2. Figure 3. Figure 4. Figure 5. Figure 6. Figure 7. Figure 8. Figure 9. Figure 10. Figure 11. LIST OF FIGURES Structures of sphingolipids. Bioactive products of Sphingolipid hydrolysis. Dietary sphingomyelin and its turnover products can reach the colon. Elution profiles of sphingosine, sphinganine, Czo-sphingosine on C1; reverse-phase HPLC. Structures of Sphingoid bases and ceramides with or without the 4,5-trans double bond. Total Sphingolipid concentrations in foods. Sphingosine, sphinganine, and ceramide inhibit growth and cause death of HT-29 human colon cancer cells. Sphingosine, sphinganine, and ceramide inhibit growth and cause death of HT-29 human colon cancer cells (photographs). Sphingosine, sphinganine, and ceramide inhibit growth and cause death of HCT-l 16 human colon cancer cells. Sphingosine, sphinganine, and ceramide inhibit growth and cause death of HCT-l 16 human colon cancer cells (photographs). Sphinganine kill HT-29 human colon cancer cells by inducing apoptosis (photographs). vi Page 11 12 22 25 31 34 35 38 39 4o I. LITERATURE REVIEW A. Colon cancer 1. Colon cancer incidence. Colon cancer is the second leading cause of cancer mortality in the United States (American Cancer Society, 1996). In the world, colon cancer is the fourth most common cancer mortality (World Cancer Research Fund, 1997). North America, Europe and Australia have high incidence of colon cancer; whereas, Central and South America, Asia and Africa are areas of low incidence. Men and women have a similar rate of incidence of colon cancer (World Cancer Research Fund, 1997). 2. Colon cancer etiology. The adenoma-carcinoma hypothesis proposed by Hill et al (1978) is now a widely accepted description of the pathogenesis of colorectal cancer. The target cells of colon carcinogenesis are crypt epithelial cells (Hill et al., 1978). According to this model, the initial colorectal lesion arises as a benign adenomatous polyp that later undergoes further disorganization of cellular and tissue phenotype. Bird er al (1987) suggested that hyperproliferation of the upper crypt cells leads to the formation of aberrant crypt foci and microadenomas. Increased proliferation and decreased differentiation of colonic epithelial cells are considered to be biomarkers for an increased risk for developing colon cancer (Lipkin, 1990). A molecular model for the adenoma-carcinoma sequence can be described by the multi-step process in which cells accumulate alterations of multiple genes that control cell growth and differentiation, resulting in the neoplastic phenotype (Vogelstein et al., 1989, Fearon er al., 1990, Fearon and Vogelstein, 1990, Kinzler et al., 1991). The genes involved in colon carcinogenesis are: mutations or loss of the APC gene (a tumor-suppressor gene) (Aaltonen er al., 1993), mutation of K-ras (a prom-oncogene) and early disorganization of DNA methylation (Feinberg and Vogelstein, 1983) and late loss of p53 (a tumor- suppressor gene). People having familial adenomatous polyposis possess a mutation in the APC gene and carry an almost 100 percent risk of colon adenocarcinoma (Aaltonen et al., 1993). Hereditary nonpolyposis colorectal cancer is a syndrome which is not easily distinguished from sporadic polyposis and cancer on physical examination. This syndrome accounts for a larger proportion of colon cancer cases than familial adenomatous polyposis (Lynch and Lynch, 1985). DNA hypomethylation is an early event in colon carcinogenesis (F einberg and Vogelstein, 1983). DNA methylation is genetically controlled and the expression of the methyl transferase gene seems to be increased in the normal mucosa of cancer patients and further increased in polyp and cancer tissue (EI-Deiry et al., 1991). Several animal studies showed that isothiocyanates, which mainly are present in cruciferous vegetables, inhibited both carcinogenesis and DNA methylation (Wattenberg, 1977, Wattenberg, 1987, Steinmetz and Potter, 1991a, & Steinmetz and Potter, 1991b). Although these observations are contradictory, both hypo- and hypermethylation of DNA are considered to be hallmarks of the early stages of the carcinogenesis. 3. Colon cancer and diet. Epidemiological studies of diet, nutrient intake and colon cancer indicate that meat consumption which reflects fat and protein intake is associated with high incidence of colon cancer (Wynder and Shigematsu, 1967, Wynder et al., 1969, Drasar and Irving, 1973, Haenszel et al., 1973, Bjelke, 1973, Armstrong and Doll, 1975, Hirayama, 1981, Heaton, 1982, Phillips and Snowdon, 1985, Potter, 1989) and fruit and vegetable consumption reflecting fiber intake is associated with a protective effect against development of colon cancer (Burkitt, 1969, Manousos et al., 1983, Stubs, 1985, Trock er al., 1990, Potter, 1990, Steinmetz and Potter, 1991a, & Steinmetz and Potter, 1991b). Bile acid metabolism and volatile fatty acids might account for the association of fat and fiber consumption with colon cancer. Fat increases bile acid production and then bowel mucosa is more exposed to toxic bile acids (Hill, 1971). In contrast, fiber binds bile acids, reduces transit time, increases stool bulk, and increases bile acids fermentation to non-toxic volatile fatty acids (Stephen and Cummings, 1983, Cummings, 1983). Other factors which might affect risk of colon cancer include physical activity (Garabrant et al., 1984, Gerhardsson er al., 1986, Slattery et al., 1988), family history (Burt er al., 1985, Bufill, 1990), and alcohol consumption (Potter et al., 1982, Tuyns er al., 1988, Longnecker, 1990, Choi and Kahyo, 1991). Lee et al (1991) showed that individuals with high levels of physical activity were at the low risk for developing colon cancer (Garabrant er al., 1984, Gerhardsson et al., 1986, Slattery et al., 1988). A family history of colon cancer is related with increased risk of colon cancer (Burt et al., 1985, Bufill, 1990). Although some studies showed that alcohol consumption (mostly beer) was related with increased risk of colon cancer, cautious interpretation of these results is recommended (Potter et al., 1982, Tuyns er al., 1988, Longnecker, 1990, Choi and Kahyo, 1991). Epidemiological studies suggest that dietary fat and specific dietary fatty acids are associated with colon cancer (Potter et al., 1993); however, this association is controversial since dietary fat is highly correlated with total energy intake. Slattery et aI r—_.__—_- d"; . (1997) evaluated these potential associations using detailed dietary intake data collected in a population-based study of 1,993 colon cancer cases and 2,410 controls in three areas of the United States. In this study population, fats added in the preparation of foods such as fried foods or bakery products or fats added to other foods at the table were responsible for one-third of the total diet fat intake. Interestingly, neither total dietary fat nor specific fatty acids were associated with risk of colon cancer afier adjusting for total energy intake, physical activity, and body size. However, fats from food preparation were associated with increased risk of colon cancer among older women, while fats from foods themselves or from additions to other foods were not. Taken together, it seems that the percentage of energy fi'om fat in the diet is not a major indicator of colon cancer risk. Several investigations suggested that soy consumption may contribute to the lower rates of breast, prostate, and colon cancer in China and Japan (Setchell er al., 1984, Barnes et a1, 1990, Adlercreutz, 1990). The isoflavone genistein, which is rich in soybeans and considered as one of the most significant plant estrogens, has been reported to protect against development of colon carcinogenesis (Akiyama et al., 1987, Ogawara er al., 1989, Teraoka et al., 1989, Bourquin er al., 1996, Thiagarajan er al., submitted). Other epidemiological studies indicate that regular consumption of fermented milk products such as yogurt may be protective against some forms of cancer (Peters er al., 1992, Karnpman et al., 1994). Some lactic acid bacteria present in fermented milk, especially Lactobacilli, are components of intestinal microflora (Bianchi Salvadori, 1986) and have protective effects against pathogenic microorganisms (Mutai and Tanaka, 1987). For example, a dietary supplement of Lactobacillus acidophilus reduced the incidence of 1,2-dirnethylhydrazine-induced colon cancer in F344 rats (Goldin and Gorbach, 1980). Also, epidemiological studies have reported that, despite the high fat intake in Finland, colon cancer incidence is lower than in other countries (Malhorta, 1977, International Agency for Research on Cancer, 1983). This may be due to Finland’s high consumption of milk, yogurt, and other dairy products. Furthermore, yogurt fractions obtained by membrane dialysis on cultured mammalian intestinal cells decreased cell proliferation in both IEC-6 and Caco-2 cells and reduced the number of IEC-6 cells in the initial growth phase (Ganjarn et al., 1997). B. Dietary sphingolipids might reduce colon carcinogenesis. l. Sphingolipids regulate cell behavior. Sphingolipids include ceramides, sphingomyelin, cerebrosides, sulfatides, and gangliosides, all of which are elaborations of a long-chain (Sphingoid) base, the most common of which is an 18-carbon compound termed sphingosine (Figure 1). Sphingosine (trans-4-sphingenine) and Sphinganine (without the 4,5-trans double bond) are the most prevalent fi'ee long-chain bases of most mammalian tissues (Merrill, 1991). Except for Sphingoid bases, ceramides, and sphingomyelin, all other sphingolipids are designated glycosphingolipids since they contain carbohydrate head groups. Glycosphingolipids include neutral glycosphingolipids and acidic glycosphingolipids. Neutral glycosphingolipids contain from one (cerebroside) to 20 or more glucose units (Makita and Taniguchi, 1985, Hakomori, 1983). Acidic glycosphingolipids contain one or more sialic acid residues (gangliosides) or monoester groups (sulfatides) (Wiegandt, 1985). Sphingolipids are located mainly in plasma membranes and related organelles including the Golgi apparatus, endosomes, and lysosomes which are functionally H \ CHZOH I"'3‘, Sphingosine H CHZOH \ W\/\/\/\/\/\/\/NH Ceramide g . H WWNH Sphlngomyelin ll NANA GLU(4<-1)GAL(4<-1)GALNAC(3<-1)GAL H | \ H20 W" Ganglloside 6"" Figure 1. Structures of sphingolipids. Abbreviations used are: NANA, N-acetyl neuraminic acid; GAL, galactose; N AC, N-acetyl; GLU, glucose. associated with cellular responses to external agents such as growth factors, cytokines, extracellular matrix proteins, neighboring cells and microbial toxins and receptors (Merrill et al., 1995b). Sphingolipids can regulate cell behavior at the surface of the cell, where they bind to extracellular ligands, such as bacterial toxins, lectins from neighboring cells, and antibodies. For instance, ganglioside GMl binds to cholera toxin and mediates cholera toxin’s effects on cells by activating adenylate cyclase (F ishman, 1982) and gangliosides GD“, Gm, and Go“, function as natural receptors for Sendai virus in host cells (Markwell er al., 1981). In addition, carbohydrate sequences present in gangliosides are recognized by monoclonal antibodies raised against murine teratocarcinomas (Solter and Knowles, 1978, Gooi et al., 1981, Kannagi er al., 1982), carcinomas of the pancreas, lung, colon and stomach (Brockhaus er al., 1982) and myeloid leukemia cells and granulocytes (Skubitz er al., 1983, Urdal et al., 1983, Magnani er al., 1984). Sphingolipids might act to form a classical receptor-ligand interaction, or to define regions of the membrane with surface characteristics that aid in receptor binding and responses, internalization, recycling, or in anchoring proteins to the cell surface (Menill er al., 1993). An example of a sphingolipid acting at the surface of the cell is the interaction between ganglioside GM, and epidermal growth factor receptor (Bremer er al., 1986, Hanai et al., 1988a, Hanai er al., 1988b). Ganglioside GM3 inhibits epidermal growth factor-stimulated growth of human epiderrnoid carcinoma cells KB and A431. The mechanism involves inhibition by ganglioside GM3 of epidermal growth factor-stimulated phosphorylation of the epidermal growth factor receptors (Bremer et al., 1986). Sphingolipids can regulate cell behavior by changes that take place in the plasma membrane and/or endosomes, where complex sphingolipids and their turnover products can stimulate or inhibit receptor kinases, responses of G proteins, protein kinases, protein phosphatases, and ion transporters (Merrill, 1991). Some sphingolipid hydrolysis products could influence both the external and internal leaflets of the plasma membrane. For example, Hope and Cullis (1987) showed that ceramides are relatively nonpolar and can cross membranes readily. In addition, sphingosine seems to undergo rapid movement across membranes in the neutral form and the hydroxyls at position 2 of the Sphingoid base backbone gives a pKa near physiological pH (Merrill et al., 1989). Sphingolipids can regulate cell behavior at intracellular areas that are sensitive to products of sphingolipid turnover. Some sphingolipids, such as lysosphingolipids, N- deacylation products of sphingolipids, and free long-chain (Sphingoid) bases, can move rapidly among membranes and might affect targets at sites distant from their locations of formation. For example, sphingosine might be liberated in the plasma membrane but affect protein kinase C in the nucleus (Hannun er al., 1986a, Hannun et al., 1986b). Sphingolipids. are also able to be converted to other bioactive compounds, such as ceramides, ceramide l-phosphate, Sphingoid bases, sphingosine l-phosphate, N- methylated sphingosines, and lysosphigolipids which can affect multiple intracellular targets (Zhang et al., 1991, Merrill et al., 1993). 2. Milk sphingomyelin might prevent the progression of adenomas to adenocarcinomas in colon cancer. Recently, sphingolipids have emerged as another component of the diet which may help to protect against development of colon carcinogenesis. Using rodents, Schmelz et a1 (1994) showed that about 12% of dietary sphingolmyelin passes through the small intestine to the colon. This finding raised the possibility that consumption of sphingomyelin may provide bioactive sphingolipid metabolites such as ceramide and sphingoid bases which could inhibit the development of colon cancer (Figure 2 and Figure 3). This hypothesis was tested in initiation-promotion studies conducted by Merrill’s group using sphingomyelin isolated from nonfat dry milk (Dillehay et al., 1994, Schmelz et al., 1996). The results showed that sphingomyelin at 0.05% of the diet inhibited formation of aberrant colonic foci in CF -1 mice treated with 1,2-dimethylhydrazine (Dillehay et al., 1994). A subsequent longer term (34 weeks) study has shown that sphingomyelin at 0.1% of the diet does not reduce the number of tumors but causes a higher percentage of adenomas and lower percentage of the more advanced adenocarcinomas (Schmelz er al., 1996). Also, the potential of synthetic sphingomyelins with saturated or unsaturated sphingoid base backbones to suppress the number of aberrant colonic foci was investigated using CF -1 mice treated with 1,2- dimethylhydrazine (Schmelz er al., 1997). In this study, the reduction of the number of aberrant colonic foci by synthetic dihydrosphingomyelin (N- palmitoyldihydrosphingomyelin) (70%, p < 0.0001) was significantly greater than by synthetic sphingomyelin (N—palrnitoylsphingomyelin) (52%, p = 0.002) and milk sphingomyelin (54%, p = 0.002). This indicates that the 4,5-trans double bond is not required for the suppression of colon carcinogenesis since synthetic dihydrosphingomyelin, which lacks the 4,5-trans double bond of the sphingoid base backbone, efficiently reduced the number of aberrant colonic foci (Schmelz et al., 1997). 3. Ceramide and sphingosine may mediate the protective effect of sphingomyelin against colon cancer. The ability of dietary sphingomyelin to reduce colon carcinogenesis may be the result of turnover of sphingomyelin to bioactive metabolites such as ceramides and sphingoid bases which play important roles in signal transduction and cell regulation (Figure 2 and Figure 3). Extracellular agonists, such as certain cytokines, growth factors, and hormones, stimulate their cell surface receptors to activate a sphingomyelinase which cleaves sphingomyelin to generate cellular ceramide (Figure 2). For example, 1a,25-dihydroxyvitamin D3 (Okazaki et al., 1989), tumor necrosis factor alpha (TNF-or) (Kim et al., 1991, Mathias et al., 1991), and y—interferon (Kim et al., 1991, Dressler et al., 1992), which are inducers of differentiation of HL-60 human leukemia cells, cause hydrolysis of sphingomyelin to form ceramide. In addition, interleukin-1 (Ballou et al., 1992, Mathias et al., 1993), dexarnethasone (Ramachandran er al., 1990), complement components (Niculescu er al., 1993), fungal macrolide brefeldin A (Linardic er al., 1992), and B-sitosterol, the main phytosterol in the diet (Awad er al., 1998), were found to induce hydrolysis of sphingomyelin in other cell lines. Cz-ceramide and other short-chain water-soluble analogues of ceramide induced differentiation in HL-60 leukemia cells which mimicked the effects of lor,25- dihydroxyvitarnin D3, tumor necrosis factor alpha (TNF-a), and y—interferon on HL-60 cells (Okazaki er al., 1990). Treatment of U937 human myeloid leukemia cells with TNF-a caused sphingomyelin hydrolysis and resulted in elevation of level of ceramide. In addition, ceramide analogs or TNF-a caused intemucleosomal DNA fragmentation, a hallmark of apoptosis in myeloid and lymphoid cells (Obeid et al., 1993). Unlike tissue necrosis, which occurs in response to severe insults and injury to cells, apoptosis involves an orderly breakdown of cells. Apoptosis is characterized with chromatin condensation followed by DNA fragmentation via the activation of an endonuclease, cytoplasmic blebbing, and condensation, and finally disintegration into dense particles called 10 H WCHZOPO3-CH2N+(CH3)3 W” Sphingomyelin Diacylglycerol Sphingomyelinase Phosphocholine MCI-I20“ \ V\/\/\/\/\/\/\/\/NH ceramide ll Ceramidase Ceramide synthase Fatty acid H CH20H + H3 Sphingosine Sphingosine kinase H CH20PO3H2 +- H3 Sphingosine-1phoephate Figure 2. Bioactive products of sphingolipid hydrolysis. Intestinal Lumen Colonic Cell Sphingomyelin l Ceramide/ Ceramide! Dihydroceramide Dihydroceramide Sphingosine/ Sphingosine! Sphinganine Sphinganine Figure 3. Dietary sphingomyelin and its turnover products can reach the colon. 12 apoptotic bodies (Michaelson, 1991). Ceramide serves as a second messenger for the action of extracellular agonists by transmitting the signal to the nucleus through multiple downstream targets such as protein kinase C (zeta isofonn), ceramide-activated protein phosphatase (CAPP), and ceramide-activated protein kinase. For example, ceramide induced early down-regulation of c-myc protooncogene (Kim et al., 1991). Ceramide activated the nuclear factor kappa B (N F-kB) in permeabilized (Schutze et al., 1992) but not in intact Jurkat T cells (Dbaibo er al., 1993). NF-kB (Schutze er al., 1992) is known to participate in the control of cell proliferation, in the dephosphorylation of the retinoblastoma gene product (pr), a tumor suppressor gene that plays an important role in cell-growth suppression and regulation of cell-cycle progression. Ceramide can act as an activator of transcription of the cyclooxygenase gene (Ballou er al., 1992). Sphingosine (F aucher et al., 1988) caused phosphorylation of the epidermal growth factor receptor on threonine 669 which was independent of protein kinase inhibition and this action of sphingosine might be associated with the conversion of sphingosine to ceramide (Goldkorn er al., 1991). Furthermore, this phosphorylation is mediated by a membrane kinase (ceramide-activated protein kinase) which is activated by TNF-or in intact and in cell-free systems (Mathias er al., 1991). Ceramide-activated protein kinase shares the substrate specificity of mitogen-activated protein kinase (MAP kinase) (Joseph et al., 1993). Ceramide activates MAP kinase (Raines er a1, 1993). Ceramide-activated protein phosphatase (CAPP) acts as a mediator of the action of ceramide. Ceramide-activated protein phosphatase (CAPP) serves as a serine/threonine protein phosphatase which is activated directly and specifically by ceramide but not by dihydrocerarnide (Dobrowsky l3 and Hannun, 1992) and is inhibited by okadaic acid (Dobrowsky er al., 1993), a tumor promotor. Sphingoid bases present in the colon may also help to protect against the development of colon cancer. Schmelz et a1 (1998) reported that both sphinganine and fumonisin B,, a mycotoxin which blocks de novo sphingolipid biosynthesis and causes accumulation of sphinganine, induce apoptosis in HT-29 human colon cancer cells. Sphingosine inhibits protein kinase C (Hannun et al., 1986a) which has been related with tumor promotion (W einstein, 1988) and blocks induction of ornithine decarboxylase by phorbol esters (Gupta et al., 1988, Enkvetchakul et al., 1989). Sphingosine also inhibits the transformation of C3H10T1/2 cells (Borek et al., 1991). Sphingosine and N- methylated derivatives inhibit growth of various human tumor cell lines in vitro (Stevens et al., 1990a) and in nude mice (Endo et al., 1991) and reduce the metastatic potential of a murine melanoma cell line (Okoshi er al., 1991). In addition, Sweeney et al (1996) showed that sphingosine and its methylated derivative N,N-dimethyl sphingosine induce apotosis in a variety of human cancer cells including CMK-7, HL-60, U-937, HRT-l8, MKN-74, and COLD-205. In contrast, primary cell cultures seem to be less susceptible to sphingosine than cancer cells. Sphingosine did not induce apoptosis in normal epithelial cells such as HUVECs or rat mesangial cells, but caused apoptosis in their transformed counterparts (Sweeney et al., 1996). Thus, sphingolipids have the possibility to inhibit carcinogenesis through many mechanisms. C. Knowledge of the types and concentrations of sphingolipids in foods is limited. Although evidence is now emerging which suggests that dietary l4 sphingolipids may protect against the development of disease, knowledge of the types and concentrations of sphingolipids in foods is limited. Sphingolipids are found in all eukaryotic and some prokaryotic cells (Merrill, 1991). However little is known about their concentrations in foods. Sphingomyelin (N-acylsphingosine-1-phosphocholine or ceramide phosphocholine) is a phospholipid located mainly in the outer leaflet of the plasma membrane of most mammalian cells (Parodi, 1997). The levels of sphingomyelin have been measured in the following foods: milk (100~200 nmol/ml) (Zeisel et al., 1986), salmon (160 nmol/g), pork and beef tissues (350~390 nmol/g) and chicken (530 nmol/g) (Blank et al., 1992). Kamath and Charles (1997) reported that the major phospholipid components of Swiss cheese whey lipid fractions were phosphatidylcholine, phosphatidylethanolamine, and sphingomyelin. Total lipids in milk are composed of 0.2 to 1.0% phospholipids. Sphingomyelin represents about 30% of total milk phospholipids and the concentration of sphingomyelin in milk was found to be about 100~200 nmol/mL (Zeisel er al., 1986). However, the concentration of sphingomyelin in milk is affected by season and the cow's stage of lactation (Parodi, 1997). Lactic acid bacteria, particularly Lacrobacillus and Bifidobacteria, are commonly used in the production of fermented milk. A study of the lipid composition of dairy starters reported that sphingomyelin is present in Lactobacillus bulgaricus and Lactobacillus acidophilus (Chand et aI. , 1992). Therefore, fermented milk products may be rich in sphingolipids. Furthermore, lactic acid bacteria such as Lactobacilli and certain components of fermented milk products have been shown to protect against the development of diseases (Malhorta, 1977, Goldin and Gorbach, 1980, International Agency for Research on Cancer, 1983, Bianchi Salvadori, 1986, Mutai and 15 Tanaka, 1987, & Ganjam er al., 1997). Since sphingosine and ceramide can induce apoptosis, the protective effect of fermented milk products against diseases may be related, in part, to the dairy products’ sphingolipid content. Many studies reported that soy foods and certain components in soy products such as genistein protect against development of colon carcinogenesis (Akiyama et al., 1987, Ogawara er al., 1989, Teraoka et al., 1989, Bourquin et al., 1996, Thiagarajan et al, submitted). Ohnishi and Fujino (1982) reported that the amount of cerebroside in soybean was about 2 umol/g. The protective effect of soy products against colon cancer may also be related to sphingolipid concentration. 16 II. OBJECTIVES The specific objectives of this research are to: A. Determine the concentrations of total sphingolipids and free sphingoid bases in soy fractions, nonfat dry milk, yogurt, and Swiss cheese. B. Determine the effects of exogenous sphingolipids on growth and death of human colon cancer cells. 17 III. MATERIALS AND METHODS Chemicals. Sphingolipids were purchased from Sigma (St. Louis, MO) and Matreya (Pleasant Gap, PA) and the other chemicals were obtained from Sigma (St. Louis, MO). Foods. Yogurt (Low fat yogurt: custard style-thick & creamy, strawberry and vanila) was obtained from Yoplait Inc. (Minneapolis, MN). Swiss cheese was from Amish Country Cheese (Linwood, MI), nonfat dry milk and ultra-pasteurized coffee cream were from Kroger Co. (Cincinnati, OH). Full fat soy flakes, soy flour, and soy protein concentrate were from Central Soya (Fort Wayne, IN). Isolated soy protein with isoflavones was from Archer Daniels Midland Co. (Decatur, IL). The yogurt, Swiss cheese, and coffee cream were lyophilized in a freeze drier (Freeze Dry/Shell Freeze System, Labconco, Kansas City, Missouri). Nonfat dry milk and full fat soy flakes (whole soybeans) and the freeze-dried pellets from yogurt, Swiss cheese, and coffee cream were powdered using a mortar and pestle. Colon cancer cell lines. The human colon adenocarcinoma cell line HT-29 and the human colon carcinoma cell line HCT-116 were purchased from American Type Culture Collection (Rockville, MD). Determination of total lipids in foods. Total lipids in nonfat dry milk, yogurt, Swiss cheese, coffee cream, full fat soy flakes (whole soybeans), soy flour, isolated soy protein, and soy protein concentrate were extracted using Chloroforrn/methanol (2:1, v/v). Chloroform/methanol (2:1, v/v) at 100 mL was added to 18 5g of dry materials and fat was solublized at 50 °C for 3 hours. Solvents and solubilized lipids were removed from substrates by Whatrnan filter paper and residues were dried at 60 °C for 24 h. The amounts of lipids were determined by gravimetric analysis. Yogurt, Swiss cheese, and coffee cream which contain large amounts of moisture were freeze-dried to remove water. The freeze-dried materials of the foods were powdered using a mortar and pestle for determination of total lipids and sphingolipids. To measure moisture content in foods, raw food materials were dried at 60 °C for 24 h in VWR Scientific 1330 F Constant Temperature Oven (Forced air or gravity) (V WR Company, Philadelphia, PA) and the results were presented in Table 1. Extraction of sphingolipids from foods for the determination of total sphingolipids. Sphingolipids were isolated by the one phase extraction method (Smith and Merrill 1995). The volume of 3 mL of chloroform/methanol (1:2, v/v) was added to the sample to be extracted. C20 sphingosine at 300 pmol as an intemal standard was added to each sample to determine the concentrations of total sphingolipids. Samples were incubated at 37°C for l h in the shaker water bath (Shaker bath, Lab-Line Instrumentals, Inc., Melrose Park, IL) at 100g. After 1 h incubation, the sample was centrifuged at 800g for 10 min (Beckrnan Instruments, Inc., Fullerton, CA) and the clear supernatant was transferred to a new tube. This step was repeated for twice. Three mL chloroform/methanol (2:1, v/v) was added to the pellet. After one hour incubation at 37°C in the shaker water bath, this extract was centrifuged and the clear supernatant was collected in the tube from the first step. This step was repeated for twice. 19 The pooled chloroform/methanol extracts from the above two steps were dried in a speed vacuum concentrator (AES 2000 Automatic Environmental Speed Vac® with Vapornet, Savant Instruments, Inc., Holbrook, NY). The dried extracts were dissolved in 1 mL of chloroform. After 1 h incubation at 37°C in the shaker water bath, the extracts were centrifuged at 800g for 10 min and clear supernatant was transferred to a new tube. One mL of chloroform was added to the pellet. After 1 h incubation at 37°C in the shaker water bath, this pellet in chloroform was centrifuged and the clear supernatant was transferred to the supernatant collected in the tubes from the last step. The pooled chloroform extracts were dried in a speed vacuum concentrator. Quantitation of total sphingolipids. After the one-phase extraction step, 2 mL of 0.5 N hydrochloric acid in methanol was added and the samples were incubated at 65°C in a dry bath (Barnstead/Thermolyne, Dubuque, IA) to hydrolyze the acyl groups. After 18 h incubation, samples were neutralized with 200 uL of 5 N ammonium hydroxide. The samples were extracted with 1 mL of chloroform and 5 mL of water. This resulted in two phases of the samples: the top phase consisted of water and methanol and the lower phase consisted of lipids in chloroform. After the samples were centrifuged, the upper aqueous phase was removed (This step was repeated for twice). The chloroform layer was passed through a column filled with sodium sulfate to remove residual water. The chloroform was evaporated using a Speed Vacuum Concentrator for about 1 h. The samples were saponified by adding 1 mL of 0.1 M potassium hydroxide 20 in methanol/chloroform (4:1, v/v) and incubating the samples for 1 h in a shaker water bath at 37°C. After the 1 h saponification, the samples were extracted with 1 mL of chloroform and 5 mL of water. This resulted in two phases of the samples: the top phase consisted of water and methanol and the lower phase consisted of lipids in chloroform. After the samples were centrifuged, the upper aqueous phase was removed (This step was repeated twice). The chloroform layer was passed through a column filled with sodium sulfate to remove residual water. The chloroform was evaporated using a speed vacuum concentrator for about 45 min. The samples were redissolved in 400 uL of mobile phase (methanol: 5 mM potassium phosphate, 87:13, v/v). The sphingoid bases in samples were derivatized with the addition of 200 uL of o-phthaldehyde in 3% borate buffer at pH 10.5. The samples were centrifuged at 10,000g for 10 min (Microcentrifuge, IEC/Micromax®) and the clear upper fraction was transferred to HPLC vials and the sphingoid bases were quantitated using High-Performance Liquid Chromatography (HPLC) (Merrill et al., 1988, Riley et al., 1994). The absolute amounts of total sphingolipids were determined by the reference to the internal standard, Czo-sphingosine. The elution profiles of sphingosine, sphinganine, and Czo-sphingosine on Cl8 reverse-phase HPLC with the solvent of methanol: 5 mM potassium phosphate, pH 7.4 (87:13) were shown in Figure 4. Sphingosine, sphinganine, and Czo-sphingosine appeared at 12.5, 17.8, and 23.1 min respectively. Quantitation of free sphingoid bases- sphingosine and sphinganine. Chloroform/methanol (l :2, v/v) at 1.9 mL was added to samples. Czo-sphingosine at 300 21 23. 125 Czo-Sphingosine 1 7.846 Sphinganine 12.535 Sphingosine Retention Time (min) Figure 4. Elution profiles of sphingosine, sphinganine, and Czo-sphingosine on C13 reverse-phase HPLC. 22 pmol was added as an internal standard to samples and samples were mixed. After the addition of 100 uL of 2 N ammonium hydroxide, the samples were incubated at 37°C in a shaker water bath for 1 h. After 1 h saponification, the samples were extracted with 1 mL of chloroform and 5 mL of water. This resulted in two phases of the samples: the top phase consisted of water and methanol and the lower phase was a mixture of lipids in chloroform. After the samples were centrifuged at 800g, the upper aqueous phase was removed (This step was repeated for twice). The chloroform layer was passed through a column filled with sodium sulfate to remove residual water. The chloroform was evaporated using a speed vacuum concentrator for about 45 min. The samples were redissolved in 400 uL of mobile phase (methanol: 5 mM potassium phosphate, 87:13, v/v). The sphingoid bases in samples were derivatized with the addition of 200 uL of o- phthaldehyde in 3% borate buffer at pH 10.5. The samples were centrifuged at 10,000g for 10 min (Microcentrifuge, IEC/Micromax®) and the clear upper fraction was transferred to HPLC vials and the sphingoid bases were quantitated using High- Perforrnance Liquid Chromatography (HPLC) (Merrill er al., 1988, Riley et al., 1994). The absolute amounts of free sphingoid bases were determined by the reference to the internal standard, Czo-sphingosine. The elution profiles of sphingosine, sphinganine, and Czo-sphingosine on Cl8 reverse-phase HPLC with the solvent of methanol: 5 mM potassium phosphate, pH 7.4 (87 :13) were shown in Figure 4. Sphingosine, sphinganine, and Czo-sphingosine were appeared at 12.5, 17.8, and 23.1 min respectively. 23 Culture of human colon cancer cells. Stock cultures of PIT-29 and HCT- 116 human colon cancer cells were grown in 100 mm culture dishes (Coming, Cambridge, MA) containing Dulbeco’s Modified Eagle Medium (DMEM) supplemented with 10% fetal bovine serum at 37°C and 5% C02. Dulbeco’s Modified Eagle Medium was purchased from Gibco BRL (Life Technologies, Gaithersburg, MD). Sphingolipid stock. Sphingosine and sphinganine were prepared as a complex with bovine serum albumin at concentrations of 104 M, 5x10‘4 M, and 10'3 M. Cz-cerarnide and Cz-dihydroceramide (cell permeable and short chain analog of naturally occurring ceramide and dihydroceramide) (Figure 5) were dissolved in ethanol as a stock solution at concentrations of 10" M, 5x104 M, 10'3 M, 2x10'3 M, and 5x10‘3 M. Total nucleic acid assay. To assess the effects of sphingolipids on cell growth and death, total nucleic acids were measured as previously described (Li et al., 1990) and used as an index of cell number. Cells were seeded at a density of 3.0 x 10’ cells/mL in 6-well dishes. HT-29 and HCT-116 cells were grown in 2 mL of Dulbeco’s Modified Eagle Medium (DMEM) with 10% fetal bovine serum for 24 or 36 h to insure that cells were in log phase before treatment with sphingolipids. After 24 or 36 h, the medium was replaced with DMEM supplemented with 1% fetal bovine serum and 20 uL of various concentrations of sphingolipids were added directly to the cell culture medium. Subconfluent cells were incubated with sphingosine, sphinganine, Cz-cerarnide, or Cz-dihydroceramide at final concentrations of 1, 5, 10, 20 and 50 uM for 0, 3, 12, 24, and 48 h. At each culture period of time, floating dead cells were removed by aspiration and the remaining live cells were rinsed with 1 mL of phosphate-buffered saline (PBS). Then 24 H CHZOH \ NI'I3"' Sphingosine H CHZOH NH3+ Sphinganine H WCHZOH \ WWW” Ceramide ll H CHZOH \ H3c\,NH Cz-Ceramide H H3CVNH Cz-dihydroceramide 8 Figure 5. Structures of sphingoid bases and ceramides with or without the 4,5-trans double bond. 25 the cells were lysed with 1 mL of 0.1 M sodium hydroxide and the total nucleic acid concentration was determined by reading absorbance of the cell lysate at 260 nm using Gene Quant-RNA/DNA Calculator (Pharmacia Biotech, Piscataway, NJ). Examination of cellular morphology. Cells were treated with sphingolipids as described above and the cells were viewed and photographed using a Nikon inverted microscope (Nikon, Garden City, NY.) fitted with a Polaroid Micro Camera (Polaroid Corporation, Cambridege, MA). Microscopic detection of apoptotic cells" To determine whether sphingoid bases and ceramide kill cells via apoptosis or via a non-apoptotic pathway, cells were incubated with sphingoid bases and ceramide at 10, 20, and 50 uM for 0, 15, 30, 60, 120, or 180 min and 12, or 24 h. Cells were collected at each time and stained with acridine orange and ethidium bromide (Mishell et al., 1980). Cells were photographed using a fluorescence microscope equipped with a camera under 10X plus 40X magnification with 400/490 nm excitation and 520 nm emission (Nikon Labophoto, Nikon Inc. Instrument Group, Garden City, N. Y.). Apoptotic and non-apoptotic cells were classified by the differences in their chromatin organization (Martin and Lenardo, 1998). Viable cells with normal nuclei appeared as containing bright green chromatin with organized structure. Viable cells with apoptotic nuclei appeared as containing bright green chromatin with highly condensed or fragmented structure. Nonviable cells with normal nuclei appeared as containing orange chromatin with organized structure. Nonviable cells with apoptotic nuclei appeared as containing orange chromatin with highly condensed or fragmented Structure. 26 Statistical analysis. Data for cell grth as influenced by sphingolipids were analyzed by two-way factorial analysis of variance (ANOVA). After application of ANOVA to the data, the significance of differences in the means of total nucleic acid content in HT-29 and HCT-116 cells between control and treatment groups at specific culture periods were evaluated by multiple comparisons using the Bonferroni method. Differences were considered significant at p < 0.05. 27 IV. RESULTS A. Sphingolipid concentrations in foods. Determination of total lipids in foods. Total lipids in foods were expressed as percent of lipids on a dry weight basis (Table 1). Coffee cream, Swiss cheese, full fat soy flakes, and yogurt were relatively high in fat (54.5, 36.7, 23.1, 16.2 % respectively). Nonfat dry milk, soy flour, isolated soy protein, and soy protein concentrate contained 1.2, 2.7, 2.2, and 0.2 % fat, respectively on a dry weight basis. Determination of sphingolipids in foods. The present study showed that most sphingolipids in foods are present as complex sphingolipids and little are present as flee sphingoid bases (Table 2). The concentrations of total sphingolipids and flee sphingoid bases (nmng of dry weight) were: nonfat dry milk (203 3: 75, 146 j; 78); yogurt (138 i 55, 1.2 i 0.3); Swiss cheese (167 i 45, 6.5 1; 7.6); full fat soy flakes (609 i 627, 2.6 i 1.1); soy flour (610 i 509, 1.6 j; 0.3); isolated soy protein (210 i 112, 2.8 i- 1.7) (mean j; SD, n=4) (Table 2 and Figure 6). Sphingosine is the predominant free sphingoid base and long-chain base backbone of total sphingolipids in foods. The percentage of flee sphingoid bases and sphingoid base backbones of food complex sphingolipids that are sphingosine and sphinganine is shown in Table 3. Sphingosine accounts for 90, 74, 91, 85, 98, and 96% of the sphingoid base backbone of total sphingolipids in nonfat dry milk, yogurt, Swiss cheese, full fat soy flakes, soy flour, and isolated soy protein, respectively (I able 3). Sphingosine accounts for 97, 69, 90, 64, 77, and 73% of flee sphingoid bases in nonfat dry milk, yogurt, Swiss cheese, full fat flake, soy flour, and isolated soy protein, respectively (Table 3). 28 Table 1. Total lipids and moisture content in foods. Lipids are expressed on a dry weight basis. Data are mean 1; SD (n=2). Foods % of lipids Water (%) Nonfatdryrnilk 1.2 1; 0.07 5.4 i 0.3 Yogurt 16.2 3; 0.21 72.9 i 0.002 Swiss cheese 36.7 i 0.16 32.8 i 6.7 Coffee cream 54.5 3; 1.35 72.2 i 0.2 Full fat soy flakes 23.1 i 0.14 3.4 i 0.4 Soy flour 2.7 j; 0.14 4.5 i 0.07 Isolated soy protein 2.2 i 0.12 7.6 i 0.02 Soy protein concentrate 0.2 i 0.14 4.4: 0.04 29 N. _ H w m md th v; H mm ON: H Hue—N m.mH m6 Nd: H M.SN 50on >8 vows—om— ? H 3 no H he 3 H N. 5% H 4.20 N2 H 2: 334 H 38 see em 2 H ed 9o H ad _._ H Nu— wdmo H Nance :v H 9% came demm 8?: H8 3 =£ 5 H 3 :3 H 3 3 H 3 3. H $2 3 H 4.2 34 H on: 386 $35 Go + N; 85 + to m6 + 36 own + mm? m.: + Qmm mNm + v.2: 20> I I I I I I _ ME + S: on + 3. SH + 2: 4.2. + 28 EN + 62 3m + 3: 5.: be 352 women EoweEam 0:08.03 83 ozone—own 258.03 E 83m panama—Em oemmowcfiam Eocham mo Bum ocgwefiqm ofimowcflam 3%; be .8 3.8.5 Ems; 5.6 £85 38.2 as; Bowie 8; £33233 amen .Avuev Om H 588 8a was mien 2303 S a no Emmoaxo 8e San— .300m 5 2095588 Bum—oweflnw .N 638. 30 1200 .L O O O 800 O) O O (nmollg of dry weight) ‘5 O Total Sphingolipids N O O Dry Yogurt Swiss Soy Soy Soy milk cheese flakes flour isolate Figure 6. Total sphingolipid concentrations in foods. Data are espressed on a dry weight basis and are mean 1 SD (n=4). 31 Table 3. Percentage of flee sphingoid bases and sphingoid base backbones of food complex sphingolipids that are sphingosine and sphinganine. Data are expressed on a dry weight basis and are mean -_i-_ SD (n=4). Total Sphingolipids Free sphingoid bases Foods (%) (%) Sphingosine Sphinganine Sphingosine Sphinganine backbone backbone Nonfat Dry Milk 89.8 i 10.7 10.2 i 10.7 96.7 i 2.7 3.3 j; 2.7 Yogurt 74.0 i 9.1 26.0 1- 9.1 69.4 3; 7.5 30.6 i 7.5 Swiss Cheese 91.2 i 4.8 8.8 _-I; 4.8 90.0 i 7.7 10.4 i 7.7 Full fat soy flake 84.5 i 12.1 15.5 i 12.1 64.4 i 22.2 35.6 i 22.2 Soy flour 98.4 j; 81.6 3.2 i 2.2 76.7 1; 4.8 23.3 -_I-_ 4.8 Isolated soy protein 96.1 i 2.0 3.9 i 2.0 73.2 i 8.0 26.8 i 8.0 32 B. Differential sensitivity of human colon cancer cells to sphingoid bases and ceramide. Sphingosine, sphinganine, and ceramide inhibit growth and cause death of H T- 29 human colon cancer cells. Based upon the concentrations of total sphingolipids that were found in milk products in this study (~138-203 nmol/g) and assuming that the average American consumes ~730g of dairy products per day (USDA, 1994, Putnam and Allhouse, 1996), the sphingolipid concentration in the colonic lumen could reach >20 uM. Cells were incubated with sphingolipids at the concentrations where may be achieved after consumption of the foods based on this estimation. To determine the effects of sphingolipids on growth and death of HT-29 human colon cancer cells, subconfluent cells were treated with sphingosine, sphinganine, C2- ceramide, or Cz-dihydroceramide and the concentration of total nucleic acids was determined as an index of cell number. The concentration of total nucleic acids in control cultures doubled over 24 to 36 hours. Sphingosine, sphinganine, and ceramide caused concentration- and time-dependent decreases in total nucleic acids of HT-29 cells. Specifically, addition of sphingosine (Figure 7A) at 20 and 50 uM significantly reduced total nucleic acid concentrations within 24 hours by 50 and 66%, respectively (p <0.05) compared to corresponding controls. Sphinganine (Figure 7B) at 10, 20 or 50 uM significantly reduced total nucleic acid concentrations within 24 hours by 55, 65, and 80%, respectively (p < 0.05) compared to corresponding controls. Cz-ceramide (Figure 7C) also caused a significant reduction in the total nucleic acid concentrations at 20 and 50 uM within 48 hours (p < 0.05). Incubation with Cz-ceramide at 50 uM for 48 h killed 33 :5: 250 i; 250 ‘5 a 8 8 200 '5 ”a 35' i 150 3 2 0 O “ < 2 2 100 3 2 ° 0 E g so 3 a '2 o n 1 r '2 o 12 24 36 12 24 36 48 Culture Period (h) Culture Period (h) 2 2 a 2 150 8 20° ° '5 5 £150 .. p E 100 E 2 .2 100 I- 2 50 2 0 0 5 g so - E - J J .2 o g o . . 12 24 36 12 24 36 48 Culture Period (h) Culture PGflOd (h) ' . S hin osine, s ' anine, and ceramide inhibit growth and cause death of HT- 12:39:6qu cpblong cancer 2:111: Subconfluent cells were cultured with sphingosrne (A), sphinganine (B), Cz-ceramide (C), and Cz-dihydrocerarnide (D) at 0 (O), 1 (O), 5 (V), 10 (V), 20 (CI), and 50 (I) uM for 3, 12, 24, or 48 h and total nucleic acrd was deterrnrned as an index of cell number. Data are flom two experiments and represent mean 3; SEM (n=8). Means at each culture period with an asterik (*) are significantly different (P < 0.05) flom the corresponding controls. 34 Control 20 uM Sphingosine 50 uM Sphingosine -- Control 20 IIM Sphinganine 50 uM Sphinganine Control 20 uM Cz-ceramide 50 uM Cz-ceramide Control 20 uM Cz-dihydroceramide 50 uM Cz-dihydroceramide Figure 8. Sphingosine, sphinganine, and ceramide inhibit growth and cause death of HT- 29 human colon cancer cells (photographs). Subconfluent cells were cultured with sphingosine (A), sphinganine (B), Cz-ceramide (C), and Cz-dihydroceramide (D) at 0, 20, and 50 uM for 24 h. 35 all cells. Unlike Cz-ceramide and the sphingoid bases, Cz-dihydroceramide did not reduce total nucleic acid concentrations (Figure 7D). Changes in cellular morphology caused by treatment of the cells with the sphingolipids for 24 h are shown in Figure 8. Sphingosine, sphinganine, and ceramide inhibit growth and cause death of [JCT-116 human colon cancer cells. Additional studies were conducted to exaxnine the effects of sphingolipids on HGT-116 human colon cancer cells. The concentration of total nucleic acids in control cultures of HCT-l 16 cells doubled over 18 to 3 6 hours. Similar to the effects on HT-29 cells, sphingosine, sphinganine, and ceratxlide caused concentration- and time-dependent decreases in total nucleic acids of HCT—ll6 cells. Specifically, addition of sphingosine (Figure 9A) at 20 and 50 11M . sig11ificantly reduced total nucleic acid concentrations within 24 h by 62 and 71%, respectively (p <0.05) compared to corresponding controls. Sphinganine (Figure 9B) at 10, 20 or 50 uM significantly reduced total nucleic acid concentrations within 24 h by 55, 77, and 93%, respectively (p < 0.05) compared to corresponding controls. Cz-cerarnide at 20 RM also caused a significant reduction in total nucleic acid concentrations within 24 hours by 33% and Cz-ceramide at 50 uM for 24 h killed all cells (Figure 9C). Unlike C2- Ceramide and the sphingoid bases, Cz-dihydroceramide did not reduce total nucleic acid c("1'23etltrations (Figure 9D). Changes in cellular morphology caused by treatment of the cells With the sphingolipids for 24 h are shown in Figure 10. Sphingoid bases and ceramide difl'erentially afl'ect death of human colon “nee!" cells. To determine whether sphingoid bases and ceramide kill cells via apoptosis or via a non-apoptotic pathway, cells were stained with acridine orange and 36 ethidium bromide and chromatin organization was examined under fluorescent light (Martin and Lenardo, 1998). Apoptotic cells were found in HT-29 and HCT-116 cells cultured with sphingosine at 20 11M and sphinganine at 10 and 20 uM for 12 and 24 h (Figure 11). 37 3 300 :- 350 a 8 B 8 250 r 5 30° ' o '6 3 250 - u :2 20° ‘ ‘2 / z 3' 200 ' /'- ‘ a 150 r g /' f, < 150 - .v * j] 3 I" .2 / * '5 10° 2 100 ~ ' é ‘a’ ‘ .. t — 5° "' z 50 F I I * s s * . * .- 0 '2 o l I n 0 12 24 36 48 0 12 24 36 48 Culture Period (h) Culture Period (h) = = 250 3 €- 5 o o 2 200 - '5 g . 35. 2. 150 - 2 2 0 < 5 2 10° “ E 2; 0 E E so - E 5 a a o I I I I o 12 24 36 4a 0 12 24 36 48 Culture Period ('1) Culture Period (h) Figure 9. Sphingosine, sphinganine, and ceramide inhibit grth and cause death of HCT-116 human colon cancer cells. Subconfluent cells were cultured with sphingosine (A), sphinganine (B), Cz-cerarnide (C), and Cz-dihydroceramide (D) at 0 (O), 1 (O), 5 (V), 10 (V), 20 (C1), and 50 (I) uM for.3, 12, 24, or 48 h and total nucleic acid was determined as an index of cell number. Data are from two experiments and represent mean 1 SEM (n=8). Means at each culture period with an asterik (*) are significantly different (P < 0.05) flom the corresponding controls. 38 Control 20 uM Sphingosine 50 uM Sphingosine Control 20 uM Sphinganine 50 uM Sphinganine Control 20 uM Cz-ceramide 50 uM Cz-ceramide Control 20 uM Cz-dihydroceramide 50 uM Cz-dihydroceramide .- Figure 10. Sphingosine, sphinganine, and ceramide inhibit growth and cause death of HGT-116 human colon cancer cells (photographs). Subconfluent cells were cultured with sphingosine (A), sphinganine (B), Cz-ceramide (C), and Cz-dihydroceramide (D) at 0, 20, and 50 uM for 24 h. 39 Figure 11. Sphinganine kill PIT-29 cells by inducing apoptosis (photographs). Subconfluent cells were cultured either without sphinganine (A), or with sphinganine (B) at 10 uM for 12 h. Cells were stained with acridine orange and ethidium bromide. The right panels show cells under phase contrast and the lefi represents cells under florescent light. 40 V. DISCUSSION Many studies suggest that specific foods and their components may protect against development of colon cancer. For example, soy foods (Akiyama et al., 1987, Ogawara et al., 1989, Teraoka et al., 1989, Bourquin et al., 1996, Hughes et al., 1997, Thiagarajan et al, submitted) as well as milk and fermented milk products (Peters et al., 1992, Karnpman et al., 1994) appear to protect against the development of colon carcinogenesis. Studies are underway to identify the component of these foods which provide the protective effects and their mechanisms of action. Recently dietary sphingolipids have gained attention for their potential to protect against the development of colon cancer (Dillehay et al., 1994, Schmelz et al., 1996, Schmelz et al., 1997); however, little is known about the types and concentrations of sphingolipids in foods. In the present study sphingolipids were extracted fi'om soy fiactions, milk, and fermented milk products and quantitated via HPLC. The concentrations of total sphingolipids in full fat soy flakes, soy flour, and isolated soy protein were ~609, 610, and 210 nmng of dry weight, respectively. Although these concentrations are lower than those previously estimated for cerebroside (2 pmol/g) in soybeans (Ohnishi and Fujino, 1982), they indicate that sphingolipids are significant constituents of soy fi'actions. Furthermore, the data suggest that the processing steps to generate soy flour from full fat soy flakes using hexane extraction do not significantly influence the concentration of total sphingolipids. The present studies also show that the concentrations of total sphingolipids in nonfat dry milk, yogurt, and Swiss cheese are ~203, 138, and 167 nmng of dry weight, 41 respectively. Assuming that sphingomyelin is the major complex sphingolipid in milk, the concentration of total sphingolipids in nonfat dry milk (203 nmol/g) is comparable with the value reported for sphingomyelin in milk (100-200 nmol/mL) (Zeisel et al., 1986). The concentrations of total sphingolipids in the fermented milk products are similar to that in nonfat dry milk suggesting that dairy starter cultures which contain sphingomyelin (Tamine and Deeth, 1980, Banwart, 1989, Chand et al., 1992, Marshall, 1993) do not contribute significantly to the total sphingolipid concentration of the fermented foods. Sphingolipids are present in soy fractions and milk products primarily as complex sphingolipids; whereas, the simpler free sphingoid bases are present at low concentrations. Moreover, sphingosine is the predominant long-chain base accounting for ~64-97% of the free sphingoid bases and ~74-98% of the sphingoid base backbone of complex sphingolipids with sphinganine accounting for the balance. Presumably dietary sphingolipids must reach the colonic tissue in order to inhibit the development of colon cancer. Schmelz et a! (1994) reported that ~88% of dietary sphingomyelin is absorbed via the small intestine. Therefore, one route that dietary sphingolipids may reach the colon is via the blood. Alternatively, the remaining 12% of dietary sphingomyelin that is not absorbed passes directly into the colonic lumen (Schmelz et al., 1994). This provides another more direct route by which dietary sphingolipids may reach the colonic cells. Based upon the concentrations of total sphingolipids that were found in milk products in this study (~138-203 nmng) and assuming that the average American consumes ~730g of dairy products per day (USDA, 42 1994, Putnam and Allhouse, 1996), the sphingolipid concentration in the colonic lumen could reach >20 uM. The mechanism by which sphingolipids inhibit colon cancer is not clear. One possibility is that complex sphingolipids such as sphingomyelin are hydrolyzed to bioactive metabolites including sphingoid bases and/or ceramides. Assuming complete hydrolysis of complex sphingolipids which reach the colon, the concentration of sphingoid bases and/or ceramides that colonic cells may be exposed to via the lumen could reach >20 uM. This concentration of sphingoid bases and/or ceramides has been shown to inhibit growth and induce differentiation and/or apoptosis in a variety of tumor cells (F aucher et al, 1988, Okazaki et al, 1990, Stevens et al, 1990a, Stevens et al, 1990b, Michaelson, 1991, Goldkom et al, 1991, Endo et al, 1991, Obeid et al, 1993, Sweeney et a1, 1996). The present study showed that sphingosine and ceramide at 20 and 50 uM and sphinganine at 10, 20, or 50 uM significantly inhibit growth and caused death of HT-29 and HGT-116 human colon cancer cells. In contrast, Cydihydroceramide which lacks the 4,5-trans double bond has no effects. These results suggest that the 4,5-trans double bond is necessary for inhibition of growth and induction of death by ceramide, but not by sphingoid bases (Figure 5). This finding is consistent with the results of previous studies which showed that short-chain ceramides caused apoptosis in many systems, while dihydroceramides which lack the 4,5-trans double bond were ineffective (Bielawska et al., 1993, Obeid, et al., 1993, Tepper et al., 1995, Sawai et al., 1995, Brugg et al., 1996, Karasavvas et al., 1996). In contrast, both sphingosine and sphinganine inhibit growth 43 and induce apoptosis in a variety of cell lines and tumor xenografis (Endo et al., 1991, Ohta et al., 1995, Jarvis et al., 1996a, Sweeney et al., 1996). Interestingly, previous studies have indicated that synthetic dihydrosphingomyelin (N- palmitoyldihydrosphingomyelin) (70%, p < 0.0001) was even more effective than synthetic sphingomyelin (N-palmitoylsphingomyelin) (52%, p < 0.002) at reducing the number of aberrant colonic foci in CF -1 mice treated with 1,2-dimethylhydrazine (Schmelz et al., 1997). Taken together with our results, this suggests that synthetic dihydrosphingomyelin (and perhaps sphingomyelin) inhibits colon carcinogenesis via turnover in the colonic lumen to the free sphingoid base (sphinganine and/or sphingosine) which may in turn inhibit growth and cause death of colon cancer cells. Alternatively, Jarvis er al (1996a) showed that a combination of ceramide and sublethal concentrations of sphingosine or sphinganine was more effective at inducing DNA fi'agmentation in human myeloid leukemia cell lines HL-6O and U937 than ceramide alone. Therefore, complex sphingolipids may inhibit colon carcinogenesis via turnover to a mixture of sphingosine, sphinganine, and ceramide. Incubation of cells with sphingosine at 20 uM and sphinganine at 10 and 20 uM for 12 h induced apoptosis; whereas, Cz-ceramide did not induce apoptosis. This finding raised a possibility of the utilization of sphingoid bases as a chemotherapeutic agent since many chemotherpeutic agents have been shown to kill susceptible cells by apoptosis (Kaufmann, 1989, Walker et al., 1991, Shinomiya et al., 1994, Havrilesky et al., 1995, Huschtscha, et al., 1996). Apoptosis is a form of programmed cell death which is either launched in response to specific stimuli such as cytokines, tumor necrosis factor a, and the fas ligand (Nagata, 1992, Smith et al., 1994), or activated in response to cell injury or stress (Gerschenson and Rotello, 1992, Michaelson, 1991). Cancer cells have been known to circumvent the normal apoptotic mechanisms to prevent their self-destruction due to their many mutations (Kerr et al., 1994, Williams, 1991). Chemotherapeutic agents which induce apoptosis appear to have less side effects since the agents can selectively kill cancer cells with no cytotoxic effects on neighboring normal tissue (Kaufmann, 1989, Walker et al., 1991, Shinomiya et al., 1994, Havrilesky et al., 1995, Huschtscha, et al., 1996). The mechanisms by sphingoid bases induce apoptosis in human colon cancer cells has not well established. Wild-type p53 protein and Bel-2 oncogene were identified as two major endogenous regulators of apoptosis (V ogelstein and Kinzler, 1992, Oren, 1992, Lane, 1992, Reed, 1995, Korsmeyer et al., 1995). Wild-type p53 protein induces cell death especially in response to DNA damaging events (V ogelstein and Kinzler, 1992, Oren, 1992, Lane, 1992). In contrast, Bel-2 oncogene show anti-apoptotic function (Reed, 1995, Korsmeyer et al., 1995). Sphingosine might affect various systems to induce apoptosis. For example, sphingosine inhibits protein kinase C (Hannun et al., 1986b), Na*/K+-A'I'Pase (Oishi et al., 1990), and phospatidic acid phosphohydrolase (Lavie and Liscovitch, 1990, Jamal et al., 1991, Mullmann et al., 1991). Sphingosine activates the epidermal growth factor receptor kinase (F aucher et al., 1988, Wedegaertner and Gill, 1989) and other sphingosine-specific kinases (Pushkareva et al., 1992). Sakakura et al (1996) showed that sphingosine induce apoptosis via down-regulation of Bcl-2 oncogene in HL-60 cells. Thus, sphingoid bases might induce apoptosis by affecting multiple downstream targets. 45 Taken together, the results indicate that sphingolipids are significant constituents of dairy products and soy fractions and that the colonic concentrations of sphingoid bases and/or ceramide which may be achieved after consumption of soy fractions and fermented milk products are sufficient to inhibit growth and cause death of human colon cancer cells. 46 References Aaltonen, L., Peltomaki, P., Leach, F., Sistonen, P., Pylkkanen, L., Mecklin, J. P., Jarvinen, H., Powell, 8., Jen, J., Hamilton, 8., Peterson, G., Kinzler, K. W., Vogelstin, B., & de la Chapelle, A. (1993) Clues to the pathogenesis of familial cancer. Science. 260: 812-816. Adlercreutz, H. (1990) Western diet and western diseases: some hormonal and biochemical mechanisms and associations. Scand. J. Clin. Lab. Invest. 50:38-23S. Akiyama, T., Ishida, J ., & Nakagawa, S. (1987) Genistein, a specific inhibitor of tyrosine -specific protein kinases. J. Biol. Chem. 262: 5592-5595. American Cancer Society (1996) Cancer Facts and Figures-1996. Armstrong, B., & Doll, R. (1975) Environmental factors and cancer incidence and mortality in different countries with special reference to dietary practices. Int. J. Cancer. 15: 617-631. Awad, A. B., Von Holtz, V., Jessie, P. C., Carol, 8. F., & Chen, Y.-C. (1998) B-sitosterol inhibits the growth of HT-29 human colon cancer cells by activating the sphingomyelin cycle. Anticancer Res. 18: 471-479. Ballou, L. R., Chao, C. P., Holness, M. A., Barker, S. C., & Raghow, R. (1992) Interleukin-1 -mediated PGE-Z production and sphingomyelin metabolism: evidence for the regulation of cyclooxygenase gene expression by sphingosine and ceramide. J. Biol. Chem. 267: 20044-20050. Banwart, G. J. (1989) Chapter 9 Useful microorganisms, In: Basic Food Microbiology (Second ed.) An Avi Book, New York. Barnes, 8., Grubbs, C., Setchell, K. D.R, & Carlson, J. (1990) Soybeans inhibit mammary tumors in models of breast cancer. In: Mutagens and carcinogenesis in the diet. Pariza, M. W., Aeschbacher, H. -U., Felton, J.S., & Sato, S.(eds) Wiley-Liss, New York. Bianchi Salvadori, B. (1986) Intestinal microflora: the role of yogurt in the equilibrium of the gut ecosystem. Int. J. Immunol. Suppl. 2: 9-18. Bielawaska, A., Crane, H. M., Liotta, D. C., Obeid, L. M., & Hannun, Y. A. (1993) Selectivity of ceramide-mediated biology. J. Biol. Chem. 268: 26226-26232. 47 Bird, R. P. (1987) Observation and quantification of aberrant crypts in the murine colon treated with a colon carcinogen: preliminary findings. Cancer Lett. 37: 147-151. Bjelke, E. (1973) Epidemiological studies of cancer of the stomach, colon, and rectum. Vol. III. Case-control study of gastrointestinal cancer in Norway. Vol. IV. Case-control study of the digestive tract cancers in Minnesota. Ann Arbor Univ. Microfilms. Blank, M. L., Cress, E. A., Smith, Z. L., & Snyder, F. (1992) Meats and fish consumed in the American diet contain substantial amounts of ether-linked phospholipids. J. Nutr. 122: 1656-1661. Borek, C., Ong, A., Stevens, V. L., Wang, E., & Merrill, A. H., Jr. (1991) Long-chain (sphingoid) bases inhibits multistage carcinogenesis in mouse C3H110T1/2 cells treated with radiation and phorbol 12-myristate 13-acetate. Proc. Natl. Acad. Sci. 88: 1953-1957. Bourquin, L. D., Lee, W. Y. K., & Bennink, M. R. (1996) Genistein and daidzein suppress colon cancer cell growth individually and additively. FASEB J. 10: A795. Bremer, E. G., Schlessinger, J ., & Hakomori, S. (1986) Ganglioside-mediated modulation of cell growth: specific effects of G.,, on tyrosine phosphorylation of the epidermal growth factor receptor. J. Biol. Chem. 261: 2434-2440. Brockhaus, M., Magnani, J. L., & Herlyn, M. (1982) Monoclonal antibodies directed against the sugar sequence of lacto-N-fucopentaose III are obtained from mice immunized with human tumors. Archs. Biochem. Biophys. 217: 647-651. Brugg, B., Michel, P. P., Agid, Y., & Ruberg, M. (1996) Ceramide induces apoptosis in cultured mesencephalic neurons. J. Neurochem. 66: 733-739. Bufill, J. A. (1990) Colorectal cancer: evidence for distinct genetic categories based on proximal or distal tumor location. Ann. Intern. Med. 113: 779-788. Burkitt, D. (1969) Related disease-related cause?. Lancet. 2: 1229-1231. Burt, R. W., Bishop, D. T., Cannon, L. A., Dowdle, M. A., Lee, R. G., & Skolnick, M. H. (1985) Dominant inheritance of adenomatous colonic polyps and colorectal cancer. New Engl. J. Med. 312: 1540-1544. Chand, R., Malan, C. P., & Srinivasan, R. A. (1992) Lipid and phospholipid composition of starter cultures. J. of Dairying Foods and Home Sciences. 11: 31-36. Choi, S. Y., & Kahyo, H. (1991) Effect of cigarette smoking and alcohol consumption in the etiology of cancer of the digestive tract. Int. J. Cancer. 49: 381-386. 48 Cummings, J. (1983) Fermentation in the human large intestine: evidence and implications for health. Lancet. 1: 1206-1209. Dbaibo, G., Obeid, L. M., & Hannun, Y. A. (1993) Tumor necrosis factor-a (TNF-a) signal transduction through ceramide: dissociation of growth inhibitory effects of TNF-a from activation of nuclear factor-k8. J. Biol. Chem. 268: 17762-17766. Dillehay, D. L., Webb, 8. K., Schmelz, E. M., & Merrill, A. H., Jr. (1994) Dietary sphingomyelin inhibits 1,2-dimethylhydrazine-induced colon cancer in CFl mice. J. Nutr. 124: 615-620. Dobrowsky, R. T., & Hannun, Y. A. (1992) Ceramide stimulates a cytosolic protein phosphatase. J. Biol. Chem. 267: 5048-5051. Dobrowsky, R. T., Kamibayashi, C., Mumby, M. C., & Hannun, Y. A. (1993) Ceramide activates heterotrimeric protein phosphatase 2A. J. Biol. Chem. 268: 15523-15530. Drasar, B. S., & Irving, D. (1973) Environmental factors and cancer of the colon and breast. Br. J. Cancer. 27: 167-172. Dressler, K. A., & Kolesnick, R. N. (1990) Ceramide l-phosphate, a novel phospholipid in human leukemia (HL-60) cells. J. Biol. Chem, 265: 14917-14921. Dressler, K. A., Mathias, S., & Kolesnick, R. N. (1992) TNF-a activates the sphingomyelin signal transduction pathway in a cell-free system. Science. 255: 1715- 1718. El-Deiry, W. S., Nelkin, B. D., Celano, P., Yen, R. W., Falco, J. P., Hamilton, 8. R., & Baylin, S. B. (1991) High expression of the DNA methlytransferase gene characterizes human neoplastic cells and progression stages of colon cancer. Proc. Natl. Acad. Sci. USA. 88: 3470-3474. Endo, K., Igarashi, Y., Nisar, M., Zhou, Q., & Hakomori, S. I. (1991) Cell membrane signaling as target in cancer therapy: inhibitory effect of N,N-dimethyl and N,N,N- trimethyl sphingosine derivatives on in vitro and in vivo growth of human tumor cells in nude mice. Cancer Res. 51: 1613-1618. Enkvetchakul, B., Merrill, A. H., Jr., & Birt, D. F. (1989) Inhibition of ornithine decarboxylase activity in mouse epidermis by sphingosine sulfate. Carcinogenesis. 10: 379-381. Faucher, M., Girones, N., Hannun, Y. A., Bell, & Davis, R. (1988) Regulation of the epidermal growth factor receptor phosphorylation state by sphingosine in A431 human epidermoid carcinoma cells. J. Biol. Chem. 263: 5319-5327. 49 Fearon, E., Cho, K., & Nigro, J. (1990) Identification of a chromosome 18q gene that is altered in colorectal cancers. Science. 247: 49—56. Fearon, E., & Vogelstein, B. (1990) A genetic model for colorectal tumorigenesis. Cell. 61: 759-767. Feinberg, A. P., & Vogelstein, B. (1983) Hypomethylation of ras oncogenes in primary human cancers. Biochem. Biophys. Res. Comm. 111: 47-54. Fishbein, J. D., Dobrowsky, R. T., Bielawska, A., Garrett, S., & Hannun, Y. A. (1993) Ceramide-mediated growth inhibition and CAPP are conserved in Saccharomyces cerevisiae. J. Biol. Chem. 268: 9255-9261. Fishman, P. H. (1982) Role of membrane gangliosides in the binding and action of bacterial toxins. J. Membr. Biol. 69: 85-97. Ganjam, L. S., Thornton, W. H., Jr., Marshall, R. T., & MacDonald, R. S. (1997) Antiproliferative effects of yogurt fractions obtained by membrane dialysis on cultured mammalian intestinal cells. J. Dairy Science. 80: 2325-2329. Garabrant, D. H., Peters, J. M., Maack, T. M., & Bernstein, L. (1984) Job activity and colon cancer risk. Am. J. Epidemiol. 119: 1005-1014. Gerhardsson, M., Norell, S. E., Kiviranta, H., Pedersen, N. L., & Ahlbom, A. (1986) Sedentary jobs and colon cancer. Am J. Epidemiol. 123: 775-780. Gerschenson, L. E., & Rotello, R. J. (1992) Apoptosis: a different type of cell death. FASEB J. 6: 2450-2455. Goldin, B. R., & Gorbach, S. L. (1980) Effect of Lactobacillus acidophilus dietary supplement on 1,2-dimethylhydrazine dihydrochloride-induced intestinal cancer in rats. JNCI. 64: 263-265. Goldkom, T., Dressler, K. A., Muindi, J., Radin, N. S., Mendelsohn, J., Menaldino, D., Liotta, D., & Kolenesnick, R. N. (1991) Ceramide stimulates epidermal growth factor receptor phosphorylation in A431 human epidermoid carcinoma cells: evidence that ceramide may mediate sphingosine action. J. Biol. Chem. 266: 16092-16097. Gooi, H., Feizi, T., Kapadia, A., Knowles, B.B., Solter, D., & Evans, J. M. (1981) Stage- specific embryonic antigen involves a-l-3-fucosy1ated type 2 blood group chains. Nature (London). 292: 156-158. 50 Gupta, A. K., Fisher, G. J., Elder, T. T., Nicholoff, B. J., & Voorhees, J. J. (1988) Sphingosine inhibits phorbol ester-induced inflammation, ornithine decarboxylase activity, and activation of protein kinase C in mouse skin. J. Invest. Dermatol. 91: 486- 491. Haenszel, W., Berg, J. W., Segi, M., Kurihara, M., & Locke, F. B. (1973) Large bowel cancer in Hawaiian Japanese. J. Natl. Cancer Inst. 51: 1765-1799. Hakomori, S. (1983) In: Sphingolipids in Biochemistry (Kanfer, J. N., & Hakomori, 8., ed.), pp. 1, Plenum, New York. Hanai, N., Dohi, T., Nores, G. A., & Hakomori, S.-I. (1988a) A novel ganglioside, De-N- acetyl-GM3 (II-3-Neu-NH-2-Lac-Cer) acting as a strong promoter for epidermal growth factor receptor kinase and as a stimulator for cell growth. J. Biol. Chem. 263: 6296- 6301. Hanai, N., Nores, G. A., MacLeod, C., Torres-Mendez, C.—R., & Hakomori, S.—I. (1988b) Ganglioside-mediated modulation of cell growth: specific effects of GM, and lyso-GM, in tyrosine phosphorylation of the epidermal growth factor receptor. J. Biol. Chem. 263: 10915-10921. Hannun, Y. A. (1994) The sphingomyelin cycle and the second messenger function of ceramide. J. Biol. Chem. 269: 3125-3128. Hannun, Y. A., & Linardic, C. M. (1993) Sphingolipid breakdown products: anti- proliferative and tumor suppressor lipids. Biochem. Biophys. Acta. 1154: 223-236. Hannun, Y. A., Loomis, C. R., & Bell, R. M. (1986a) Protein kinase C activation in mixed micells. J. Biol. Chem. 261: 7184-7190. Hannun, Y.A., Loomis, C. R., & Merrill, A. H. Jr. (1986b) Sphingosine inhibition of protein kinase C activity and of phorbol dibutyrate binding in vitro and in human platelets. J. Biol. Chem. 261: 12604-12609. Harvrilesky, L. J., Elbendary, A., Hurteau, J. A., Whitaker, R. S., Rodriguez, G. C., & Berchuck, A. (1995) Chemotherapy-induced apoptosis in epithelial ovarian cancers. Obstet. Gynecol. 85: 1007-1010. Heaton, K. W. (1982) The colon and bile-acid metabolism. In: Colon and Nutrition (Kasper, H., & Goebell, H., eds.), pp. 117-125. MTP Press Ltd., Lancaster. Hill, M. J. (1971) Fecal steroid composition and its relationship to cancer of the large bowel. J. Pathol. 104: 129-139. 51 Hill, M. J., Morson, B. C., & Bussey, H. J. R. (1978) Aetiology of adenoma-carcinoma sequence in large bowel. Lancet. 1: 245-247. Hirayama, T. A. (1981) A large-scale cohort study on the relationship between diet and selected cancers of the digestive organs. In Gastrointestinal Cancer: Endogenous Factors (Bruce, W. R., Correa, P., Lipkin, M., Tannenbahurn, S. R., & Wikins, T. D., eds.), Banbury Report No. 7, pp. 409-429, CSH, New York. Hope, M., J., & Cullis, P. R. (1987) Lipid asymmetry induced by transmembrane pH gradients in large unilamellar vesicles. J. Biol. Chem. 262: 4360-4366. Hughes, J. S., Ganthavom, C., & Wilson-Sanders, S. (1997) Dry beans inhibit azoxymethane-induced colon carcinogenesis in F344 rats. J. Nutr. 127: 2328-2333. Huschtscha, L. I., Bartier, W. A., Ross, C. E. A., & Tattersall, M. H. N. (1996) Characteristics of cancer cell death after exposure to cytotoxic drugs in vitro. Br. J. Cancer. 73:54-60. lntemational Agency for Research on Cancer, Intestinal Microecology Group (1983) Dietary fiber, transit-time, fecal bacteria, steroids and colon cancer in two Scandinavian populations. Lancet. 2: 207-211. Jamal, 2., Martin, A., Munoz, A. G., & Brindley, D. N. (1991) Plasma membrane function from rat liver contains a phosphatidate phosphohydrolase distinct from that in the endoplasmic reticulum and cytosol. J. Biol. Chem. 266: 2988-2996. Jarvis, W. D., Fornari, F. A., Traylor, R. S., Martin, H. A., Kramer, L. B., Erukulla, R. K., Bittman, R., & Grant, 8. (1996a) Induction of apoptosis and potentiation of ceramide- mediated cytotoxicity by sphingoid bases in human myeloid leukemia cells. J. Biol. Chem. 271: 8275-8284. Jarvis, W. D., Grant, 8., & Kolesnick, R. N. (1996b) Ceramide and the induction of apoptosis. Clin. Cancer Res. 2: 1-6. Joseph, C. K., Byun, H.-S., Bittman, R., & Kolesnick, R. N. (1993) Substrate recognition by ceramide-activated protein kinase: evidence that kinase activity is proline-directed. J. Biol. Chem. 268: 20002-20006. Kamath, S. R, & Charles, V. M. (1997) Recovery and composition of Swiss cheese whey lipid-containing fractions. J. Agric. Food Chem. 45: 4271-4274. Karnpman, E., Goldbohm, R. A., Van den Brandt, P. A., & Van’t veer, P. (1994) Fermented dairy products, calcium, and colorectal cancer in the Netherlands cohort study. Cancer Res. 54: 3186-3190. 52 Kannagi, R., Nudelman, E., Levery, S. B., & Hakomori, S. (1982) A series of human erythrocyte glycosphingolipids reacting to the monoclonal antibody directed to a developmentally regulated antigen, SSEA-l. J. Biol. Chem. 257: 14865-14874. Karasavvas, N., Erukulla, R. K., Bittman, R., Lockshin, R., & Zakeri, Z. (1996) Stereospecific induction of apoptosis in U937 cells by N-octanoyl-sphingosine stereoisomers and N-octyl-sphingosine. Eur. J. Biochem. 236: 729-737. Kaufmann, S. (1989) Induction of endonucleolytic DNA cleavage in human acute myelogenous leukemia cells by etoposide, camptothecin, and other cytotoxic anticancer drugs: a cautionary note. Cancer Res. 49: 5870-5879. Kerr, J. F. R., Winterford, C. M., & Harmon, B. V. (1994) Apoptosis: its significance in cancer and cancer therapy. Cancer. 7 3: 2013-2026. Kim, M. Y., Linardic, C., Obeid, L., & Hannun, Y. (1991) Identification of sphingomyelin turnover as an effector mechanism for the action of tumor necrosis factor a and y—interferon. J. Biol. Chem. 266: 484-489. Kinzler, K., Nibert, M., & Vogelstein, B. (1991) Identification of a gene located at chromosome 5q21 that is mutated in colorectal cancers. Science. 251: 1366-1370. Korsmeyer, S. J., Yin, X. M., Oltvai, Z. N., Veis-Novack, D. J., & Linette, G. P. (1995) Reactive oxygen species and the regulation of cell death by the Bcl-2 gene family. Biochim. Biophys. Acta. MoI. Basis. Dis. 1271: 63-66. Lane, D. P. (1992) p53, guardian of the genome. Nature. 358: 15-26. Lavie, Y., & Liscovitch, M. (1990) Activation of phospholipase D by sphingoid bases in NG 108-15 neural-derived cells. Biochem. Biophys. Res. Comm. 167: 607-613. Lee, I. M., Paffenbarger, R. 8., Jr., & Hsieh, C. (1991) Physical activity and risk of developing colorectal cancer among college alumni. J. Natl. Cancer Inst. 83: 1324- 1329. Li, I. -C., Chang, C. C., & Trosko, J. E. (1990) Thymidylate synthetase gene as a quantitative mutation marker in Chinese hamster cells. Mutat. Res. 243: 233-239. Linardic, C. M., Jayadev, S., & Hannun, Y. A. (1992) Brefeldin A promotes hydrolysis of sphingomyelin. J. Biol. Chem. 267: 14909-14911. Lipkin, M. (1990) Biomarkers of increased susceptibility to gastrointestinal cancer: new applications to studies of cancer prevention in human subjects. Cancer Res. 50: 5761- 5760. 53 Longnecker, M. P. (1990) A case-control study of alcoholic beverage consumption in relation to risk of cancer of the right colon and rectum. Cancer Causes Control. 1: 5-14. Lowry, O. H., Rosebrough, N. J., Farr, A. L., & Randall, R. J. (1951) Protein measurement with the folin phenol reagent. J. Biol. Chem. 193: 265-275. Lynch, P. M., & Lynch, H. T. (1985) In: Colon cancer genetics. New York, NY, Van Nostrand Rheinhold. Magnani, J., Ball, E. E., Franger, M. W., Hakomori, S., & Ginsburg, V. (1984) Monoclonal antibodies PMN 6, PMN 29 and PM-8l bind differently to glycolipids containing a sugar sequence occurring in lacto-N-fucopentaose III. Archs. Biochem. Biophys. 233: 501-506. Makita, A., & Taniguchi, N. (1985) In: Glycolipids (Wiegandt, H., ed.), pp. 1. Elsevier, New York. Malhorta, S. L. (1977) Dietary factors in a study of colon cancer fi'om cancer registry, with special reference to the role of saliva, milk and fermented milk products and vegetable fiber. Med Hypotheses. 3: 122-126. Manousos, 0., Day, N. E., Trichopoulos, D., Gerovassilis, F., Tzonou, A., & Polychronopoulou, A. (1983) Diet and colorectal cancer: a case-control study in Greece. Int. J. cancer. 32:1-5. Markwell, M. A. K., Svennerholm, L., & Paulson, J. C. (1981) Specific gangliosides function as host cell receptors for Sendai virus. Pro. Natl. Acad. Sci. USA. 78: 5406- 5410. Marshall, V. M. (1993) Starter cultures for milk fermentation and their characteristics. J. Society of Dairy Technol. 46: 49-56. Martin, D., & Lenardo, M. (1998) Microscopic quantitation of apoptotic index and cell viability using vital and fluorescent dyes. In: Current Protocols in Immunology. (Coligan, J. E., Kruisbeek, A. M., Margulies, D., Shevach, E. M., & Strober, W. eds.) John Wiley & Sons, Inc. Mathias, S., Dressler, K. A., & Kolesnick, R. N. (1991) Characterization of a ceramide- activated protein kinase: stimulation by INF-a. Pro. Natl. Acad Sci. USA. 88: 10009- 10013. 54 Mathias, 8., Younes, A., Kan, C.-C., Orlow, L., Joseph, C., & Kolesnick, R. N. (1993) Activation of the sphingomyelin pathway in intact EL4 cells and in a cell-free system by IL-lB. Science. 259: 519-522. Merrill, A. H. Jr. (1991) Cell regulation by sphingosine and more complex sphingolipids. J. Biomemb. Bioenerget. 23: 83-104. Merrill, A. H., Jr., Hannun, Y. A., & Bell, R. M. (1993) Introduction: sphingolipids and their metabolites in cell regulation. Adv. Lip. Res. 25: 1-24. Merrill, A. H., Jr., Lingrell, S., Wang, E., Nikolova-Karakashian, M., Vales, T. R., & Vance, D. E. (1995a) Sphingolipid biosynthesis de novo by rat hepatocytes in culture. J. Biol. Chem. 270: 13834-13841. Merrill, A. H., Jr., Nimkar, S., Menaldino, D., Hannun, Y. A., Loomis, C., Bell, R. M., Tyagi, S. R., Lameth, J. D., & Stevens, V. L. (1989) Structural requirements for long- chain (sphingoid) base inhibition of protein kinase C in vitro and for the cellular effects of these compounds. Biochemistry. 28: 3138-3145. Merrill, A. H., Jr., Schemelz, E. M., Dillehay, D. L., Spiegel, S., Shayaman, J. A., Schroeder, J. J., Riley, R. T., Voss, K. A., & Wang, E. (1997) Sphingolipids-The enigmatic lipid class: biochemistry, physiology, and pathophysiology. Toxicol. Appl. Pharm. 142: 208-225. Merrill, A. H., Jr., Schmelz, E. M., Wang, E., Schroeder, J. J., Dillehay, D. L., & Riley, R. T. (1995b) Role of dietary sphingolipids and inhibitors of sphingolipid metabolism in cancer and other diseases. J. Nutr. 125: 16778-16828. Merrill, A. H., Jr., Wang, E., Mullins, R., Jaminson, W., Nimkar, S., & Liotta, D. (1988) Quantitation of free sphingosine in liver by high performance liquid chromatography. Anal. Biochem. 171: 373-381. Messina, M., Persky, V., Setchell, K., & Barnis, S. (1994) Soy intake and cancer risk: A review of the in vitro and in vivo data. Nutr. Cancer. 21: 113-131. Michaelson, J. (1991) In: Apoptosis: the molecular basis of cell death (T omei, L. D., & Cope, F. 0., eds) pp. 31-46, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY. Mishell, B. B., Shiigi, S. M., Henry, C., Chan, E. L., North, J., Gallily, R., Slomich, M., Miller, K., Marbrook, J., Parks, D., & Good, A. H. (1980) Preparation of mouse cell suspensions. In: Selected methods in cellular immunology (Mishell, B. B., & Shiigi, S. M. eds.) pp. 21-23. W. H. Freeman, New York. 55 Mullmann, T. J ., Siegel, M. 1., Egan, R. W., & Billah, M. M. (1991) Sphingosine inhibits phosphatidate phosphohydrolase in human neutrophils by a protein kinase C-independent mechanism. J. Biol. Chem. 266: 2013-2016. Mutai, M., & Tanaka, R. (1987) Ecology of Bifidobacteria in the human intestinal microflora. Bifidobactria Microflora. 6: 33-41. Nagata, S. (1994) Apoptosis regulated by a death factor and its receptor: Fas ligand and Fas. Phil. Trans. R. Soc. Lond Biol. 345: 281-287. Niculescu, F., Rus, H., Shin, S., Lang, T., & Shin, M. L. (1993) Generation of diacylglycerol and ceramide during homologous complement activation. J. Immunol. 150: 214-224. Obeid, L. M., Linardic, C. M., Karolak, L. A., & Hannun, Y. A. (1993) Programmed cell death induced by ceramide. Science. 259: 1769-1771. Ogawara, H., Akiyama, T., & Watanabe, S. (1989) Inhibition of tyrosine protein kinase activity by synthetic isoflavones and flavones. J. Antibiot. 42: 340-343. Ohnishi, M., & Fujino, Y. (1982) Sphingolipids in immature and mature soybeans. Lipids. 17: 803-810. Ohta, H., Sweeney, E. A., Masamune, A., Yatomi, Y., Hakomori, S-I., & Igarashi, Y. (1995) Induction of apoptosis by sphingosine in human leukemic HL-60 cells: a possible endogenous modulator of apoptotic DNA fragmentation occurring during phorbol ester- induced differentiation. Cancer Res. 55: 691-697. Oishi, K., Zheng, B., & Kuo, J. F. (1990) Inhibition of Na”/I("-ATPase and sodium pump by protein kinase C regulators sphingosine, lysophosphatidylcholine, and oleic acid. J. Biol. Chem. 265: 70-75. Okazaki, T., Bell, R. M., & Hannun, Y. A. (1989) Sphingomyelin turnover induced by vitamin D3 in HL-60 cells. J. Biol Chem. 264: 19076-19080. Okazaki, T., Bielawska, A., Bell, R. M., & Hannun, Y. A. (1990) Role of ceramide as a lipid mediator of 1a, 25-dihydroxy vitamin D3-induced HL-60 cell differentiation. J. Biol. Chem. 265: 15823-15831. Okoshi, H., Hakomori, 8., Zhou, Q., Kimura, S., Tashiro, K., & Igarahi, Y. (1991) Cell membrane signaling as target in cancer therapy: 11. Inhibitory effect of N,N,N-trimethyl sphingosine on metastatic potential of murine Bl6 melanoma cell line through blocking of tumor cell-dependent platelet aggregation. Cancer Res. 51: 6019-6024 56 Oren, M. (1992) The ultimate tumor suppressor gene?. FASEB J. 6: 3169-3176. Parodi, P. W. (1997) Cow's milk fat components as potential anticarcinogenic agents. J. Nutr. 127: 1055-1060. Peters, R. K., Pike, M., C., Garabrant, D., & Mack, T. M. (1992) Diet and colon cancer in Los Angeles County, California. Cancer Causes Control. 3:457-473. Phillips, R. L., & Snowdon, D. A. (1985) Dietary relationships with fatal colorectal cancer among Seventh-day Adventists. J. Natl. Cancer Inst. 74: 307-317. Potter, J. D. (1989) Large bowel cancer: epidemiology and biology. Findings in Gastraenteralagy. 24: 137-140. Potter, J. D. (1990) The epidemioloy of fiber and colorectal cancer: why don’t the epidemiologic data make better sense?. In: Dietary Fiber (Kritchevsky, D., Bonfield, C., & Anderson, J. W., eds.) pp. 431-436. Plenum Press, New York. Potter, J. D., McMichael, A. J., & Hartshome, J. M. (1982) Alcohol consumption and beer consumption in relation to cancers of bowel and lung: an extended correlation analysis. J. Chronic. Dis. 35: 833-842. Potter, J. D., Slattery, M. L., Bostick, R. M., & Gapstur, S. M. (1993) Colon cancer: a review of the epidemiology. Epidemiol. Rev. 15: 499-545. Pushkareva, M. Y., Wasiuddin, A. K., Alessenko, A. V., Sahyoun, N., & Hannun, Y. A. (1992) Sphingosine activation of protein kinases in Jurkat T cells: in vitro phosphorylation of endogenous protein substrates and specificity of action. J. Biol. Chem. 267: 15246-15251. Putnam, J. J., & Allshouse, J. E. (1996) Food consumption, prices, and expenditures. Annual Data Published by the United States Department of Agriculture. Raines, M. A., Kolesnick, R. N., & Golde, D. W. (1993) Sphingomyelinase and ceramide activate mitogen-activated protein kinase in myeloid HL-60 cells. J. Biol. Chem. 268: 14572-14575. Ramachandran, C. K., Murray, D. K., & Nelson, D. H. (1990) Dexamethasone increases neutral sphingomyelinase activity and sphingosine levels in 3T3-L1. Biochem. Biophy. Res. Commun. 167: 607-613. Reed, J. C. (1995) Bel-2: prevention of apoptosis as a mechanism of drug resistance. Hematol. Oneal. Clirt North. Am. 9: 451-473. 57 Riley, R. T., Wang, E., & Merrill, A. H., Jr. (1994) Liquid chromatographic determination of sphinganine and sphingosine: Use of the free sphinganine-to- sphingosine ration as a biomarker for consumption of furnonisins. JA OAC. 77: 533-540. Sakakura, C., Sweeney, E. A., Shirahama, T., Hakomori, S., & Igarashi, Y. (1996) Suppression of Bel-2 gene expression by sphingosine in the apoptosis of human leukemic HL-60 cells during phorbol ester-induced terminal differentiation. FEBS Lett. 379: 177- 180. Sawai, H., Okazaki, T., Yarnamoto, H, Okano, H., Takeda, Y., Tashima, M., Sawada, H., Okuma, M., Ishikura, H., Umehara, H., & Domae, N. (1995) Requirement of AP—I for ceramide-induced apoptosis in human leukemia HL-60 cells. J. Biol. Chem. 270: 27326- 27331. Schmelz, E. M., Bushnev, A. S., Dillehay, D. L., Liotta, D. C., & Merrill, A. H., Jr. (1997) Suppression of aberrant colonic crypt foci by synthetic sphingomyelins with saturated or unsaturated sphingoid base backbones. Nutr. Cancer. 28: 81-85. Schmelz, E. M., Crall, K. J., Larocque, R., Dillehay, D. L., & Merrill, A. H., Jr. (1994) Uptake and metabolism of sphingolipids in isolated intestinal loops. J. Nutr. 124: 702- 712. Schmelz, E. M., Dillehay, D. L., Webb, S. K., Reiter, A., Adams, J., & Merrill, A. H., Jr. (1996) Sphingomyelin consumption suppresses aberrant colonic crypt foci and increases the proportion of adenomas versus adenocarcinomas in CF] mice treated with 1,2- dimethylhydrazine: implications for dietary sphingolipids and colon carcinogenesis. Cancer Res. 56: 4936-4941. Schmelz, E. M., Dombrink-Kurtzrnan, M. A., Roberts, P. C., Kozutsumi, Y., Kawasaki, T., & Merrill, A. H., Jr. (1998) Induction of apoptosis by fumonisin Bl in HT-29 cells is mediated by the accumulation of endogenous free sphingoid bases. T axical. Appl. Pharm. 148: 252-260. Schutze, S., Potthoff, K., Macheidt, T., Berkovic, D., Wiegrnann, K., & Kronke, M. (1992) TNF activates NF-kB by phosphatidylcholine-specific phospholipase C-induced acidic sphingomyelin breakdown. Cell. 71: 765-776. Setchell, K. D. R., Borriello, S. P., Hulme, P., Kirk, D. N., & Axelson, M. (1984) Nonsteroidal estrogens of dietary origin: possible roles in hormone-dependent disease. Am. J. Clin. Nutr. 40: 569-578. Shinomiya, N., Shinomiya, M., Wakiyama, H., Katsura, Y., & Rokutanda, M. (1994) Enhancement of CDDP cytotoxicity by caffeine is characterized by apoptotic cell death. Exp. Cell. Res. 210: 236-242. 58 Skubitz, K. M., Pressano, S., Bottero, L., Ferreo, D., Rovera, G., & August, A. J. (1983) Human granulocyte surface molecules identified by murine monoclonal antibodies. J. Immunol. 131: 1882-1888. Slattery, M. L., Potter, J. D., Duncan, D. M., & Berry T. D. (1997) Dietary fats and colon cancer: assessment of risk associated with specific fatty acids. Intl. J. Cancer. 73: 670- 677. Slattery, M. L., Schumacher, M. C., Smith, West, D. W., & Abd-Elghany, N. (1988) Physical activity, diet and risk of colon cancer in Utah. Am J. Epidemiol. 128: 989-999. Smith, C. A., Farrah, T., & Goodwin, R. G. (1994) The TNF receptor superfamily of cellular and viral proteins: activation, costimulation, and death. Cell. 76: 959-962. Smith, E. R., & Merrill, A. H., Jr. (1995) Differential roles of de nova sphingolipid biosynthesis and turnover in the burst of free sphingosine and sphinganine, and their 1- phosphates and N-acyl derivatives that occurs upon changing the medium of cells in culture. J. Biol. Chem. 270: 18749-18758. Salter, D., & Knowles, B. B. (1978) Monoclonal antibody defining a stage-specific embryonic antigen (SSEA-l). Proc. Natl. Acad Sci. USA. 75: 5565-5569. Steinmetz, K. A. & Potter, J. D. (1991a) Vegetables, fruit, and cancer. I. Epidemiology. Cancer Causes and Control. 2: 325-357. Steinmetz, K. A. & Potter, J. D. (1991b) Vegetables, fi'uit, and cancer. 11. Mechanisms. Cancer Causes and Control. 2: 427-442. Stephen, A., & Cummings, J. (1983) Mechanism of action of dietary fiber in the human colon. Nature. 284: 283-284. Stevens, V. L., Nimkar, S., Jamison, W. C. L., Liotta, D. C., & Merrill, A. H., Jr. (1990a) Characteristics of the growth inhibition and cytotoxicity of long-chain (sphingoid) bases for Chinese hamster ovary cells: evidence for and involvement of protein kinase C. Biochem. Biophys. Acta. 1051: 37-45. Stevens, V. L., Owens, N. E., Winton, E. F., Kinkade, J. M., Jr., & Merrill, A. H., Jr. (1990b) Modulation of retinoic acid-induced differentiation of human leukemia (I-IL60) cells by serum factors and sphinganine. Cancer Res. 50: 222-226. Stubs, P. (1985) The anatomy of abuses. London, pp. 61. In: Royal College of Physicians: Medical Aspects of Dietary Fiber. Pitrnan Medical, Bath, 1980. Sweeney, E. A., Sakakura, C., Shirahama, T., Masamune, A., Ohta, H., Hakomori, S-I., & Yasuyuki, J. (1996) Sphingosine and its metylated derivative N, N-dimethlysphingosine 59 (DMS) induce ap0ptosis in a variety of human cancer cell lines. Int. J. Cancer. 66: 358- 366. Tamine, A. Y., & Deeth, H. C. (1980) Yogurt: technology and biochemistry. J. Food Prat. 43: 939. Tepper, C. L., Jayadev, S., Lin, B., Bielawska, A., Wolff, R. A., Yonehara, S., Hannun, Y. A., & Seldin, M. F. (1995) Role for ceramide as an endogenous mediator of fas- induced cytotoxicity. Proc. Natl. Acad. Sci. USA. 92: 8443-8447. Teraoka, H., Ohmura, Y., & Tsukada, K. (1989) The nuclear matrix from rat liver is capable of phosphorylating exogenous tyrosine-containing substances. Biochem. Intern. 18: 1203-1210. Thiagarajan, D. G., Bennink, M. R., Bourquin, L. D., & Kavas, F. A. (Submitted) Prevention of precancerous colonic lesions in rats by say flakes, soy flour, genistein, and calcium. Am. J. Clin. Nutr. Track, 8., Lanza, E., & Greenwald, P. (1990) Dietary fiber, vegetables, and colon cancer: critical review and meta analyses of the epidemiologic evidence. J. Natl. Cancer Inst. 82: 650-661. Tuyns, A. J ., Kaaks, R., & Haelterrnan, M. (1988)Colorecta1 cancer and the consumption of foods: a case control study in Belgium. Nutr. Cancer. 11: 189-204. Urdal, D. L., Brentnall, T. A., Bernstein, I. D., & Hakomori, S. I. (1983) A granulocyte reactive monoclonal antibody, lGlO, identifies the GalBl-4(Fucorl-3)-GlcNAc (X determinant) expressed in HL-60 cells on both glycolipid and glycoprotein molecules. Blood. 62: 1022-1026. Vander, A., Sherman, J. H., Luciano, D. S. (1994) Chapter 17 The digestion and absorption of food, In: Human Physiology (Six ed.) pp. 567, 574-575, 592. McGraw- Hill, Inc., N.Y. Vogelstein, B., Fearon, E., & Kern, S. (1989) Allelotype of colorectal carcinomas. Science. 244: 207-212. Vogelstein, B., & Kinzler, K. W. (1992) p53 function and dysfunction. Cell. 70: 523- 526. Walker, P. R., Smith, C., Youdale, T., Leblanc, J., Whitfield, J. F., & Sikorska, M. (1991) Topoisomerase II-reactive chemotherpeutic drugs induce apoptosis in thymocytes. Cancer Res. 51: 1078-1085. 60 Wattenberg, L. W. (1977) Inhibition of carcinogenic effects of polycyclic hydrocarbons by benzyl isothiocyanate and related compounds. J. Natl. Cancer Inst. 58: 195-198. Wattenberg, L. W. (1987) Inhibition of chemical carcinogenesis. J. Natl. Cancer Inst. 60: 11-18. Wedegaertner, P. B., & Gill, G. N. (1989) Activation of the purified protein tyrosine domain of the epidermal growth factor receptor. J. Biol. Chem. 264: 11346-11353. Weinstein, I. B. (1988) The origins of hrunan cancer: molecular mechanisms of carcinogenesis and their implications for cancer prevention and treatrnent-twenty-seventh G. H. A. clows memorial award lecture. Cancer Res. 48: 4135-4143. Wiegandt, H. (1985) In: Glycalipids (W iegandt, H., ed.), pp. 199. Elsevier, New York. Williams, G. T. (1991) Programmed cell death: apoptosis and oncogenesis. Cell. 65: 1097-1098. World Cancer Research F und/American Institute for Cancer Research (1997) Chapter 4.10 Colon, Rectum. In: F aad, Nutrition and the Prevention of Cancer: a global perspective. pp. 218-251. American Institute for Cancer Research. Wynder, E. L., & Shigematsu, T. (1967) Environmental factors of cancer of the colon and rectum. Cancer. 20: 1529-1561. Wynder, E. L., Kajitani, T., Ishikana, S., Dodo, H., & Tako, A. (1969) Environmental factors of cancer of the colon and rectum. 11. Japanese epidemiologic data. Cancer. 23:1210-1220. Zeisel, S. H., Char, D., & Sheard, N. F. (1986) Choline, phosphatidyl choline and sphingomyelin in human and bovine milk and infant formulas. J. Nutr. 116: 50-58. Zhang, H., Desai, N. N., Olvera, A., Seki, T., Brooker, G., & Spiegal, S. (1991) Sphingosine-l-phosphate, a novel lipid, involved in cellular proliferation. J. Cell Biol, 114: 155-167. 61 HICHIGQN srnTE UNIV. LIBRRRIES illiliWIilHllWW W" H W "W 1| W "I” W WI 31293 17743950 1 (3