rlllllllllllllWi. (a. ‘..' ; LIBRARY aid-29791 Michigan State University This is to certify that the dissertation entitled Chemopreventive and chemotherapeutic mechanisms of sphingoilpid metabolites in human colon cancer cells and breast stem, normal, and tumorigenic cells presented by Eun Hyun Ahn has been accepted towards fulfillment of the requirements for the PhD. degree in Human Nutrition- Environmental ToximflL Major Professor's Signature I?- 4 ~a3 Date MSU is an Alfinnetive Action/Equal Opportunity Institution PLACE IN RETURN BOX to remove this checkout from your record. TO AVOID FINES return on or before date due. MAY BE RECALLED with earlier due date if requested. DATE DUE DATE DUE DATE DUE 6/01 c:/ClRC/DateDua.p65-p.15 Chemopreventive and chemotherapeutic mechanisms of sphingolipid metabolites in human colon cancer cells and breast stem, normal, and tumorigenic cells By Eun HyunAhn A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY in Human Nutrition-Environmental Toxicology Department of Food Science & Human Nutrition and Institute of Environmental Toxicology 2003 ABSTRACT CHEMOPREVENTIVE AND CHEMOTHERAPEUTIC MECHANISMS OF SPHINGOLIPID METABOLIT ES IN HUMAN COLON CANCER CELLS AND BREAST STEM, NORMAL, AND TUMORIGENIC CELLS By Eun Hyun Ahn The effects of the sphingolipid metabolites (sphingosine, sphinganine, ceramide, and dihydroceramide) on proliferation and differentiation in human normal epithelial cells versus tumorigenic cells and stem cells have not previously been compared. This dissertation research addressed these questions using following cell culture models: HT - 29 and HCT-l 16 human colon cancer epithelial cells, human breast epithelial cells (HBEC) developed from tissues of healthy women obtained during reduction mammoplasty (type I HBEC with stem cell characteristics as target cells for breast carcinogenesis and type H HBEC with basal epithelial cell phenotypes as breast normal cells) and transformed tumorigenic type I HBEC as breast cancer cells. The objectives of the present study were to: I) investigate the effects of sphingoid bases (sphingosine and sphinganine), ceramide, and dihydroceramide on proliferation, cell cycle, and apoptosis of HT-29 and HGT-116 cells; 2) determine the effects of sphinganine on major isoforms of mitogen activated protein kinases (ERK, JNK, and p38) and AKT (protein kinase B) in HT-29 cells; 3) evaluate the effects of sphingoid bases on proliferation, cell cycle, and apoptosis of tumorigenic type I HBEC and type I HBEC compared to type II HBEC; and 4) determine the effects of sphingoid bases on the ability of type I HBEC to differentiate to type H HBEC. Data fiom HT—29 and HGT-116 cells indicate that sphingosine, sphinganine, and Cz-ceramide inhibited the growth and arrested cell cycle at Gz/M phase. Sphingosine, sphinganine, and Cz-ceramide at 20—50 uM induced apoptosis, with sphinganine being the most potent of the metabolites studied. In contrast, Cz-dihydroceramide had no effect, suggesting that the 4,5-trans double bond is necessary for the inhibitory effects of Cz-ceramide, but not for inhibitory effects of sphingoid bases. Sphinganine at an apoptosis-inducing concentration (35 uM) caused early and strong activation of JNKZ/JNKI and p38 and early inhibition of AKT, with minimal effects on activation of ERKl/ERKZ in HT-29 cells. Results fi'om HBEC show that sphinganine more potently inhibited the growth and induced apoptosis of tumorigenic type I I-[BEC than sphingosine (ICso for sphinganine 4 uM; sphingosine 6.4 pM). Sphingoid bases (8-10 uM) arrested cell cycle at G2/M with reductions in S and 60/6. phases. Sphinganine also more potently inhibited the growth and caused death of type I HBEC than sphingosine, while the same concentrations of both sphinganine and sphingosine had minor effects on type II HBEC. At concentrations (0.05-0.5uM), which are below the growth inhibitory range, sphingoid bases induced differentiation of type I HBEC to type II HBEC, as detected with concomitant expression of the novel tumor suppressor protein, maspin, in type II HBEC. In conclusion, sphingoid bases and ceramide might possess chemotherapeutic properties against colon cancer, with sphinganine being the most potent. Activation of JNK and p38 and inhibition of AKT might mediate sphinganine-induced apoptosis of colon cancer cells. Data indicate the potential for sphingoid bases to be employed as chemotherapeutic and chemopreventive agents against human breast cancer. Dedicated to my mother Sun Yul Yu, my late father Byung Hyun Ahn and my older brothers Suk Yul Ahn and Hyoung You] Ahn who show endless love toward me and God who created the world and human beings and continuously teaches us to love one another. iv ACKNOWLEDGEMENT I sincerely thank my precious advisor Dr. Joseph J. Schroeder for his capable advise, warm encouragement, guidance, and patience throughout my Ph.D. program. He is always willing to spend his time and share his life for his graduate students. He has shown me what it means to walk with maturity and to show genuine care toward others and mutual respect under any circumstances. He has taught me to work professionally while pursuing a team spirit and practicing kindness and hospitality. All of these nice and excellent aspects are the attributes I wish to pass unto the next generation. I am grateful to my mentor and Ph. D. guidance committee Dr. Chia-Cheng Chang for his excellent and clear advice, insights on science, faithfulness, thoughtfulness, composure, and patience. I was fortunate to research with human breast epithelial cells (stem cells, normal cells, and tumorigenic cells) Dr. Chang’s group has developed. While I worked at Dr. Chang’s lab for nine months, I finished the last part of my second research project of colon cancer and started and completed the last breast cancer project. Dr. Chang was available day and night to provide close and professional advice. I was honored to learn how to culture human breast epithelial cells directly from Dr. Chang. I thank my academic advisor Dr. Dale R. Romsos for his timely advice, support, patience, and wisdom. In the midst of his busy schedule, he spent time with me on a regular basis during the last stage of my Ph.D. program, which helped me to focus on my research and to plan my career after the graduation. He provided helpful suggestions and objective perspectives on my breast cancer project. I thank Drs. Maurice R. Bennink and Leslie D. Bourquin who were members of my Ph. D. guidance committee for helpful comments and encouragement. I am grateful to Dr. Brad Upham for participating in my Ph.D. dissertation defense evaluation process and for his helpful suggestions and warm smile. My appreciation extends to Dr. James E. Trosko for his encouragement and support. It was a great opportunity for me to get to know Dr. Trosko during the last period of my Ph.D. program. I thank for their technical advice in various areas the following: Drs. Louis King and Zahidul Islam on flow cytometry analysis, Dr. Youngjoo Chung on western blotting, Dr. Ludmila Roze and Rebecca Uzarski on fluorescence microscopic detection of apoptotic cells, and Therese Soderdahl on immunofluorescence staining. Drs. Chung and Islam’s friendship made my journey of Ph.D. program more joyful. I thank Drs. James Pestka and John Linz for providing access to their experimental instruments. I thank Michelle Komosinski and Mark Hagerty for excellent technical assistance on quantitation of colonies of human breast epithelial cells. I am grateful to fellow graduate students Min Sun Kim, Eun Hee Lee, Chi Zhang, and Hong Yang for their companionship, fiiendship, and enthusiasm of science while we worked together in the same laboratory during my Ph.D. and MS. program. I thank people (Maki Satoh, Gina Brooks, Therese Soderdahl, Dr. Mei-Hui Tai, Chad Coe, Shauna Matlen, Andrea Satoh, and short-term visiting scholars) at Drs. Chang, Trosko, and Upham’s lab for their companionship and fi'iendship. I thank previous and/or current staff at our dept (Mary Schneider, Debie Klein, Shelli Pfeifer, and Sharon Hart for their service. I thank Dr. Won 0. Song for her loving encouragement and advice since I came to MI, USA. I appreciate Dr. Mun Shig Son at University of Vermont for his father-like vi encouragement. I thank fellow graduate students at our department (Dr. loo-Won Lee, Seung-Yeon Lee, Dr. Micheal Miller, Dr. Soc-Young Kang, Yikyung Park, Chia-F en Chung, Jean Kerver, and Sungyong Hong, Dr. Hanseung Shin, Dr. Sangjin Chung, Saori Obayashi, Dr. Miyoung Jang, and many others) for fi'iendship. I thank my best fiiends J ungae Nam and Mia Chung, my relatives (J ieun Kim, Bockyul You, Woosung Lee, Ingoo Lee, Hyungoo Lee, Keumyul you, Youhae Kim, J ihoon Kim, Soonyul You, and many others), my precious and close fiiends Dr. Hae- Sook Park and Susan Park Song, and sincere fi'iends at University Reformed Church in East Lansing, MI (Dr. Irvine Widders, Romelia Widders, Gary Brinkman, Jaeun Joo, Kisoon Kim, Stacey Bieler, Dr. Tom Bieler, and pastor Tom Stark), and nice and old fi'iends Yeonsook Lim, Yoonyoung Yang, and Oakja Back for encouragement. I am grateful to my American host family (Alice, Ian, and Stanely Morgan and Tory, Dale, Benett Foster) for their prayer in love. I thank the beautiful children with special needs at the Lansing Area Parent’s Respite Center, MI for the time we shared together. I thank Dr. David A. Talmage at Columbia University for his great patience, encouragement, and support during my final stage of dissertation. vii TABLE OF CONTENTS LIST OF TABLES LIST OF FIGURES LIST OF ABBREVIATIONS CHAPTER 1. Introduction Rationale of this study Objective and hypothesis CHAPTER 2. Literature review A. Cancer A.1. Cancer as a disease of proliferation and differentiation A.2. Cancer through mutations A.3. Environmental factors and cancer B. Colon cancer 8.]. Colon cancer incidence B.2. Colon cancer etiology and risk factors 32.]. Colon carcinoma development B.2.2. Family history as a risk factor for colon cancer B.2.3. Genes associated with high risk for colon cancer B.2.4. Other risk factors for colon cancer 33. Dietary factors in colon cancer B.3.l . Dietary fat B.3.2. Caloric restriction B.3.3. Dairy products B.3.4. Soy products C. Breast cancer C. l . Breast cancer incidence C.2. Breast cancer etiology and risk factors C.2.1. Family history as a risk factor for breast cancer C.2.2. Genes associated with high risk for breast cancer C.2.3. Other risk factors for breast cancer C.3. Dietary factors in breast cancer C.3.1. Dietary fat C.3.2. Caloric restriction C.3.3. Dairy products C.3.4. Soy products viii page xiii xiv xix UuNtd 10 11 11 12 14 14 14 14 18 22 22 22 26 D. Cancer chemotherapy and chemoprevention D. l . Chemotherapy D.1.l. Definition of chemotherapy D. 1 .2. Goals for use of chemotherapy D.2. Chemoprevention D.2.l . Definition of chemoprevention D.2.2. Characteristics of the chemopreventive agents D.2.3. Defense mechanisms for damaged cells D.3. Induction of apoptosis by chemotherapeutic and/or chemopreventive agents D.3.1. Removal of unwanted cells via apoptosis D.3.2. Induction of apoptosis by chemotherapeutic agents D.3.3. Induction of apoptosis by chemopreventive agents D.4. Roles of stem cell differentiation for cancer prevention D.4.l . Characteristics of stem cells D.4.2. Differentiation genes D.4.3. A mechanism of CAMP-induced differentiation D.5. Suppression of telomerase activity as a novel chemotherapy D.5.1. Telomeres D.5.2. Telomerase D.5.3. High telomerase activity in cancers D.5.4. Roles of telomerase activity on proliferation and differentiation D.5.5. Suppression of telomerase activity as a novel chemotherapy D.5.6. Regulation of telomerase E. Sphingolipids as possible chemotherapeutic and chemopreventive agents E.l. Sphingolipids structures E.2. Sphingolipid signaling pathway: Ceramide generation caused by sphingomyelinase activation E.3. Sphingolipids occurrence and dietary sphingolipids E.4. Prevention of colon carcinogenesis by dietary sphingomyelin E.4.1. Uptake and metabolism of sphingolipids E.4.2. Inhibition of colon carcinogenesis by sphingolipids in rodent models E.5. Inhibition of human breast cancer cell growth by sphingolipids E.6. Ceramide and sphingoid bases as mediators for various anticarcinogenic agents E.7. Induction of apoptosis by ceramide and sphingoid bases E.8. Regulation of apoptosis by Bel-2 family genes E.9. Regulation of apoptosis by mitogen activated protein kinases (MAPKs) E.9.l. Major isoforms of MAPKs (ERK, JNK, and p38) E.9.2. Roles of ERK, JNK, and p38 E.9.3. Activations of ERK, JNK, and p38 E.9.4. Apoptosis triggered by imbalance between ERK, JNK, and p38 E.9.5. Regulation of MAPKs by sphingolipids 3O 30 31 33 35 37 41 41 42 45 45 47 48 49 49 5 l E.10. Regulation of apoptosis by AKT E.10.1. AKT activation as cell survival and anti-apoptotic signal E.10.2. Activation/phosphorylation of AKT at thr308 and ser473 E.10.2. Regulation of AKT by sphingolipids E.11. Involvements of calcium or calmodulin in proliferation and apoptosis E.12. Summary: Mechanisms of actions of sphingolipids in proliferation, apoptosis, and differentiation F. Cellular models for chemotherapy and chemoprevention studies F .1. Human colon cancer cells RI .1. HT-29 cells F.1.2. HCT-116 cells F .1 .3. HT-29 and HCT-l 16 cells for chemotherapy studies F.2. Human breast epithelial cells (HBEC) F .2.]. Type I HBEC as normal stem cells F.2.2. Transformed tumorigenic type I HBEC F.2.3. Type II HBEC as normal cells F.2.4. HBEC for chemotherapy and chemoprevention studies F.2.5. Criteria for identification of chemotherapeutic and chem0preventive agents in HBEC 56 59 61 62 62 CHAPTER 3. Sphingoid bases and ceramide induce apoptosis in HT-29 and HCT-ll6 human colon cancer cells A. ABSTRACT B. INTRODUCTION C. MATERIALS AND METHODS C. l . Chemicals and reagents C.2. Sphingolipid treatment C.3. Cell culture C.4. Assessment of cell proliferation C.5. Fluorescence microscopic detection of apoptotic cells C.6. Isolation and quantitation of fiagmented DNA C.7. Flow cytometric analysis of cell cycle and population D. RESULTS D. l . Sphingosine, sphinganine, and Cz-ceramide inhibit growth and cause death of HT-29 and HCT-116 human colon cancer cells D.2. Fluorescence microscopic detection of apoptotic cells in HT-29 and HCT-l 16 cells D.3. Quantitation of fragmented DNA using diphenylamine assay D.4. Flow cytometric analysis of cell cycle of HT—29 and HGT-116 cells E. DISCUSSION CHAPTER 4. Sphinganine causes early activation of ERK, JNK, and p38 and early inhibition of AKT activation in HT-29 human colon cancer cells A. ABSTRACT B. INTRODUCTION C. MATERIALS AND METHODS C.1. Sphinganine preparation C.2. Cell culture C.3. Isolation and quantitation of cellular protein C.4. Western blot analysis for protein expression of ERKl/ERKZ, JNK2/JNK1, p3 8, and AKT and phosphorylated forms of ERKl/ERKZ, JNK2/JNK1, p38, AKT-ser473, and AKT-thr308 C.5. Densitometry D. RESULTS D. 1 . Sphinganine causes early activation of ERKl/ERK2 D.2. Sphinganine causes early activation of JNK2/JNK1 D.3. Sphinganine causes early activation of p38 MAP kinase D.4. Sphinganine causes early inhibition of AKT-ser473 and AKT- thr308 E. DISCUSSION xi 72 73 75 79 84 95 103 104 105 108 111 117 CHAPTER 5. Evaluation of sphinganine and sphingosine as breast cancer 121 chemotherapeutic and chemopreventive agents using human breast stem, normal, and tumorigenic cell models A. ABSTRACT 122 B. INTRODUCTION 124 C. MATERIALS AND METHODS 129 CI. C2. C3. C.4. C5. C6. C7. Q8. Q9. Sphingolipids HBEC culture media Development of human breast epithelial cells (HBEC) from reduction mammoplasty tissues of healthy females Separation of type I HBEC with stem cell characteristics and type II HBEC Derivation of in vitro neoplastically transformed HBEC lines from HBEC, normal human mammary epithelial culture fiom reduction mamoplasty tissues Culture of type I HBEC, tumorigenic type I HBEC, and type H HBEC Assessment of cell proliferation Flow cytometry of cell cycle and population Assessment of type I HBEC differentiation C.10. Immunofluorescence staining C.11. Statistical analyses D. RESULTS D.1. D2. D3. D4. D5. D6. 137 Sphinganine more potently inhibits the growth and causes death of tumorigenic type I HBEC than sphingosine Sphinganine more potently induces apoptosis of tumorigenic type I HBEC than sphingosine. Both sphingoid bases arrest cell cycle at Gle with reductions in S and Go/Gl phases. Sphinganine more potently inhibits the growth and causes death of type I HBEC than sphingosine Type II HBEC are less sensitive to growth inhibitory effects of sphinganine and sphingosine than tumorigenic type I HBEC and type I HBEC Sphinganine and sphingosine induce differentiation of type I HBEC to type H HBEC Sphinganine increases the expression of a tumor suppressor protein, maspin during induction of differentiation of type I stem HBEC to type II HBEC E. DISCUSSION 154 CHAPTER 6. APPENDIX Conclusion and significance of the study 162 168 REFERENCES l 82 xii LIST OF TABLES Page CHAPTER 2 Table 2.1. Modes of cell death: Distinct features of necrosis and 34 apoptosis Table 2.2. Different effects of W7 on apoptosis induced by various agents 60 in a variety of cells Table 2.3. The differences in gene mutation status in HT-29 and HCT- 65 116 cells Table 2.4. Phenotypic differences between type I and type H human 71 breast epithelial cells (HBEC) Table 5.1. Sphingoid bases induce differentiation of type I HBEC 151 (HME29-A) to type II HBEC APPENDIX Table A. 1 . Mobility of sphingolipids on Thin-Layer Chromatography 169 Table A2. Rfvalues of sphingolipids on 2-Dimemsional Thin-Layer 169 Chromatography xiii Figure 2.1. Figure 2.2. Figure 2.3. Figure 2.4. Figure 2.5. Figure 2.6. Figure 2.7. LIST OF FIGURES CHAPTER 2 Multi-step model for the colon carcinoma development Sphingolipids signaling pathway: Ceramide and sphingosine as mediators for protective effects of sphingomyelin against cancers Mitogen Activated Protein (MAP) Kinases signaling pathways The cell survival and anti-apoptotic actions of AKT HT-29 and HGT-116 human colon cancer cells. Type I normal human breast epithelial cells (HBEC), tumorigenic type I HBEC, and type H HBEC (photographs) Differentiation of type I HBEC with stem cell characteristics to type H HBEC and formation of ductal and terminal end bud- like structure fiommixture oftypelandtype H HBEC (photographs) Derivation of tumorigenic type I HBEC and type 11 normal human breast epithelial cells (HBEC) from type I HBEC with stem cell characteristics xiv Page 55 58 69 70 Figure 3.1. Figure 3.2. Figrue 3.3. Figure 3.4. Figure 3.5. Figure 3.6. Figure 3.7. Figure 3.8 CHAPTER 3 Structures of sphingosine, sphinganine, ceramide, Cz-ceramide, and Cz-dihydroceramide Effects of sphingoid bases and ceramides on growth and death of HT-29 and HOT-116 human colon cancer cells Effects of sphingoid bases and ceramides on chromatin and nuclear condensation in HT-29 and HCT -1 16 human colon cancer cells (photographs) Effects of sphingoid bases and ceramide on fragmented low molecular DNA in HT -29 and HCT-116 human colon cancer cells Sphingosine increases the number of sub-Go/G. cells indicative of apoptosis (one representative histogram) Effects of sphingoid bases and ceramides on HT-29 and HCT- 116 apoptotic cell number Effects of sphingoid bases and ceramides on cell cycle distribution of HT-29 and HGT-116 human colon cancer cells Regulation of cell growth, cell cycle, and apoptosis by sphingoid bases and ceramides in PIT-29 and HCT-l 16 hrnnan colon cancer cells Page 78 85 87 89 92 93 101 Figure 4.1. Figure 4.2. Figure 4.3. Figure 4.4. Figure 5.1. Figure 5.2. Figure 5.3. Figure 5.4. Figure 5.5. Figure 5.6. Figure 5.7. Figure 5.8. CHAPTER 4 Effects of sphinganine on activation of extracellular signal- regulated kinase 1/2 (ERKl/ERKZ) in HT-29 human colon cancer cells Sphinganine causes early activation of c-Jun NH2-terminal kinase-2/1 (JNK2/JNK1) in HT-29 human colon cancer cells Sphinganine causes early activation of p38 mitogen activated protein kinase in HT-29 human colon cancer cells Sphinganine causes early inhibition of AKT-ser473 and AKT- thr308 in HT-29 human colon cancer cells. CHAPTER 5 Structures of sphingosine, sphinganine, ceramide, C2- ceramide, and Cz-dihydroceramide Derivation of tumorigenic type I HBEC and type H normal human breast epithelial cells (HBEC) from type I HBEC with stem cell characteristics Sphinganine more potently inhibits the growth and causes death of tumorigenic type I HBEC than sphingosine Sphinganine more potently inhibits the growth and causes death of tumorigenic type I HBEC than sphingosine at 24 h (photographs) Sphinganine increases the number of A0 (sub-GO/Gl) cells, indicative of apoptosis, and arrests cell cycle at G2/M phase in tumorigenic type I HBEC (one representative histogram) Sphinganine more potently induces apoptosis of tumorigenic type I HBEC than sphingosine. Both sphingoid bases at high concentrations (8-10 uM) arrest cell cycle at G2/M phase with reductions in S and Go/G1 phases Effects of sphinganine and sphingosine on growth and death of type I HBEC (HME29-A) and type H HBEC (HME29-A) Sphinganine inhibits the growth and causes death of type I HBEC (HME30-4A) (photographs) xvi 113 114 115 116 126 127 138 139 142 143 145 146 Figure 5.9. Figure 5.10. Figure 5.11. Figure 5.12. Figure A.1. Figure A.2. Figure A.3. Figure A.4. Figure A.5. Figure A.6. Figure A.7. Type H HBEC (HME29-A) are less sensitive to growth inhibitory effects of sphinganine and sphingosine than tumorigenic type I HBEC and type I HBEC at 6 (1 (photographs) Sphinganine and sphingosine induce differentiation of type I HBEC (HME29-A) to type II HBEC (Representative photographs) Sphinganine increases the expression of a tumor suppressor protein maspin during induction of differentiation of type I HBEC (HME29-A) to type H HBEC (photographs) Chemopreventive properties of sphinganine and sphingosine in type I and type H HBEC and chemotherapeutic effects of sphinganine and sphingosine against tumorigenic type I HBEC and type I HBEC APPENDIX Effects of exogenous Crceramide and Cz-dihydroceramide on intracellular concentrations of free sphingoid bases in HCT- 1 16 human colon cancer cells Effects of sphinganine (SA) on gap junctional intercellular communication on HOT-116 human colon cancer cells (photographs) Effects of sphinganine on gap junctional intercellular commrmication on HT-29 human colon cancer cells (photographs) Effects of sphingosine, sphinganine, Cz-ceramide, and C2- dihydroceramide on DNA fragmentation, indicative apoptosis, in HT-29 human colon cancer cells Effects of sphingosine, sphinganine, Cz-ceramide, and C2— dihydroceramide on DNA fragmentation, indicative apoptosis, in HGT-116 human colon cancer cells Effects of sphingoid bases on apoptotic cell number in HCT- 116 human colon cancer cells Effects of sphingoid bases on cell cycle distribution of HCT- 116 human colon cancer cells xvii 147 152 153 171 171 172 173 174 175 175 Figure A.8. Effects of a calmodulin antagonist W7 on growth inhibitory 176 effects of sphinganine (SA) in HCT-116 human colon cancer cells Figure A.9. Effects of sphinganine and sphingosine on gap junctional 178 intercellular communication on trunorigenic type I HBEC at 24 h (photographs) Figure A.10. Sphinganine (SA) more potently inhibits the growth and 179 causes death of tumorigenic type I HBEC than sphingosine at 2 d (photographs) Figure A.11. Effects of sphinganine and sphingosine on cell cycle 179 distribution in tumorigenic type I HBEC at 12h Figure A.12. Effects of bovine serum albumin (BSA) on growth of 180 tumorigenic type I HBEC and type H HBEC Figure A.13. Fibroblasts and the third type of cells are found in human 180 breast epithelial cells (HME23A, HME29A, HME30-4A, HME3 l-L2A) (photographs) Figure A. 14. Type II HBEC (HME29A) are less sensitive to growth 181 inhibitory effects of sphinganine and sphingosine than tumorigenic type I HBEC and type I HBEC at 3 (1 (photographs) xviii HBEC PHSZ COX2 FBS HNPCC APC FAP AFAP FCC PCR AP-l CRE CREB PKA NSAID EGF PDGF TNFor IL-l RTK MEK MEKK INK/SAPK cPLA2 MSKI CHOP p90RSK MAPKs ERK p38 BMK-l/ERKS MEK kinase PBK PPARr LISTS OF ABBREVIATIONS 3’, 5’-cyclic Adenosine Monophosphate Human Breast Epithelial Cells Prostaglandin H synthase 2 Cyclooxygenase 2 Fetal Bovine Serum Hereditary Nonpolyposis Colorectal Cancer Adenomatous Polyposis Coli Familial Adenomatous Polyposis Attenuated form of Familial Adenomatous Polyposis Familial Colorectal Cancer Polymerase Chain Reaction Activator protein-1 c-AMP response element c-AMP response element binding protein Protein kinase A NOnsteroidal anti-inflammatory drugs Eidermal Gowth Factor Patelet Derived Growth Factor Tumor Necrosis Factor-or Interleukin-1 Receptor Tyrosine Kinase MAP kinase kinase MAP/ERK kinase MAP/ERK kinase kinase c-Jun NHz-terminal Kinase/Stress Activated Protein Kinase JNK kinase cytosolic phopholipase A2 mitogen-and stress-activated protein kinase-1 cAMP response element binding protein homologous protein Ribosomal p90 S6 kinase Mitogen Activated Protein kinases Extracellular signal-Regulated Kinase p3 8/RK/C SBP Big Mitogen-activated protein Kinase-l/ERKS Phosphatidylinositol 3-kinase Peroxisome Proliferator Activated Receptor gamma xix CHAPTER 1. INTRODUCTION Rationale of this study In recent years, sphingolipids have emerged as components of the diet which may help to protect against development of colon carcinogenesis (for review: Vesper et al. 1999, Dillehay et al. 1994, Schmelz et al. 1994, Schmelz et al. 1996, Schmelz et al. 1997, Schmelz et al. 1998, Schmelz et al. 2000, Schmelz et al. 2001). Complex sphingolipids are important components of dairy and soy products (Ahn and Schroeder, 2002a), two classes of foods that appear to have the potential to prevent carcinogenesis and some other diseases. Sphingolipid digestion products such as ceramide and sphingoid bases (sphingosine and sphinganine) play important roles in regulating cell proliferation, differentiation, and apoptosis and have been implicated as putative second messengers in signaling pathways (Merrill et al. 1997). This raises the possibility that complex sphingolipids may protect against carcinogenesis via digestion to ceramide or sphingoid bases. Schmelz et al (1994) reported that ~88% of dietary sphingomyelin is digested and absorbed as ceramides and sphingoid bases in the small intestine and small amounts of dietary sphingolipids appear in lymph (Nilsson, 1968, Nilssorr, 1969) and serum (Imaizumi et al. 1992). Thus, the colon and other tissues could be exposed to ceramide and sphingoid bases via the circulation. Alternatively, ~12% of dietary sphingomyelin is not absorbed and passes directly into the colonic lumen where it may be metabolized by colonic bacteria. This provides a more direct route by which colonic tissues may be exposed to ceramide and sphingoid bases. Evidence is emerging that bioactive digestion products of sphingolipids also may have the potential to both prevent and treat human breast cancer. Yang et al (submitted) reported that sphingosine kills tumorigenic breast cells by inducing apoptosis (programmed cell death) while not affecting type H normal human breast epithelial cells (HBEC). These findings strengthen the potential use of sphingolipids as chemotherapeutic agents since this form of programmed cell death can kill target tumor cells without influencing adjacent normal cells (Sweeney et al. 1996). In addition, Yang et al (Submitted) found that low concentrations of sphingosine caused differentiation of type I HBEC (having stem cell characteristics) to type H HBEC suggesting that sphingosine may also have chemopreventive properties by reducing the number of stem cell targets of carcinogenesis. Although recent studies suggest that dietary sphingolipids may protect against the development of colon cancer and possibly breast cancer, the mechanism by which sphingolipids kill colon and breast cancer cells is not clear. One possibility is that sphingolipids alter the expression of genes that modulate apoptosis. For example, ceramide induces apoptosis in PC-12 cells via the activation of caspase-3 and these effects were blocked by overexpression of bcI-2 suggesting that ceramide may activate caspase-3 by down-regulating bcl-Z (Yoshimura et al. 1998). Alternatively, apoptosis induced by sphingosine and ceramide in other cell-types is accompanied by an altered balance between extracellular signal-regulated kinase (ERK), c-Jun NHz-terminal kinase/stress activated protein kinase (JNK/SAPK), and p38 mitogen activated protein kinases (MAPKs) (Xia et al. 1995, Kummer et al. 1997, Jarvis et al. 1997). In addition, the AKT pathway is a probable target because ceramide (Schubert et a1, 2000) and sphingosine inhibit phosphorylation (activation) of AKT (a cell survival signal) to induce apoptosis in human liver cancer cells (Chang et al, 2001b). The mechanism by which sphingoid bases cause differentiation of type I HBEC to type II HBEC (which are more resistant to carcinogenesis) is unclear. Other agents that trigger this differentiation elevate cAMP. Therefore, sphingoid bases could activate adenylate cyclase, inhibit phosphodiesterase activity, or act via a cAMP-independent mechanism. In the present studies, HT-29 and HCT-116 human colon cancer cells and transformed tumorigenic type I HBEC will be used to examine mechanisms by which sphingolipids inhibit growth and cause death of colon and breast cancer cells. Also, effects of sphingolipids on growth of type H HBEC representing normal breast cells will be tested. Type I HBEC will be used to investigate the mechanism by which sphingoid bases stimulate differentiation of breast cells to type H HBEC. Objective and hypothesis The overall objective of this proposed research is to investigate roles of sphingolipid metabolites sphingoid bases (sphingosine and sphinganine), Cz-ceramide, and Cz-dihydroceramide on cell growth, apoptosis (a programmed cell death), cell cycle, differentiation, and cell signaling pathways in HT—29 and HGT-116 human colon cancer cells and type I human breast epithelial cells (HBEC), type H HBEC, and transformed tumorigenic type I HBEC. The specific aims of the research are to: Study 1: Sphingoid bases and ceramide induce apoptosis in HT-29 and HCT- 116 human colon cancer cells (CHAPTER 3) Aim 1. Determine effects of sphingosine, sphinganine, Cz-ceramide, and C2- dihydroceramide on growth and death of HT-29 and HGT-116 human colon cancer cells. Aim 1 Hypothesis. Since complex sphingolipids inhibit colon carcinogenesis in rodent models, sphingosine, sphinganine, and Cz-ceramide will inhibit growth and cause death of HT-29 and HCT-116 human colon cancer cells. Aim 1 Null Hypothesis. Cz-dyhydroceramide will inhibit growth and cause death of HT- 29 and HCT-116 human colon cancer cells. Aim 2. Determine whether sphingosine, sphinganine, Cz-ceramide, and C2- dihydroceramide cause death of HT-29 and HCT-l 16 human colon cancer cells by inducing apoptosis. Aim 2 Hypothesis. Sphingosine, sphinganine, and Cz-ceramide will cause death of HT-29 and HCT-116 human colon cancer cells by inducing apoptosis. Aim 2 Null Hypothesis. Cz-dyhydroceramide will cause death of HT-29 and HCT-116 human colon cancer cells by inducing apoptosis. Aim 3. Determine whether sphingosine, sphinganine, Cz-ceramide, and C2- dihydrocerarrride inhibit growth of HT-29 and HGT-116 human colon cancer cells by arresting the cell cycle at a specific phase. Aim 3 Hypothesis. Sphingosine, sphinganine, and Cz-ceramide will inhibit growth of HT-29 and HGT-116 human colon cancer cells by arresting the cell cycle at a specific phase. Aim 3 Null Hypothesis. Cz-dihydroceramide will inhibit growth of HT-29 and HCT-116 human colon cancer cells by arresting the cell cycle at a specific phase. Study 2: Sphinganine causes early activation of JNK and p38 and early inhibition of AKT in HT-29 human colon cancer cells (CHAPTER 4) Aim 4. Determine effects of sphinganine on mitogen activated protein kinases (MAPKs), including extracellular signal-regulated kinase (ERK), c-Jun NHz-terminal kinase/stress-activated protein kinase (JNK/SAPK), and p3 8, and AKT that are responsible for regulating cell proliferation and apoptosis in HT-29 human colon cancer cells. Aim 4.A. Determine the time required for an apoptotic concentration of sphinganine to affect active phosphorylated forms of MAPKs (ERKl/ERK2, JNK2/JNK1, and p38) and the expression levels of MAPKs in HT-29 cells. Aim 4.A. Hypothesis. Since MAPKs play significant roles in regulating cell proliferation and apoptosis, sphinganine will alter phosphorylation status of MAPKs (ERKl/ERK2, JNK2/JNK1, and p38) in HT-29 cells. Aim 4.B. Determine the time required for an apoptotic concentration of sphinganine to affect activation of AKT by phosphorylation at ser473 and thr3 08 and the expression levels of AKT in HT-29 cells. Aim 4.B. Hypothesis. Since AKT plays significant roles in regulating cell proliferation and apoptosis, sphinganine will alter phosphorylation of AKT at ser473 and thr308 in HT-29 cells. Study 3: The study of chemotherapeutic and chemopreventive roles of sphinganine and sphingosine in breast carcinogenesis using human breast stem, normal, and'tumorigenic cell models (CHAPTER 5) Type I human breast epithelial cells (HBE C) with stem cell characteristics Aim 5.A. Determine effects of sphinganine on growth and death of type I HBEC in comparison to sphingosine. Aim 5.A. Hypothesis. Sphinganine will inhibit growth and cause death of type I HBEC to a similar degree as the known cytotoxic effects of sphingosine. Aim 5.B. Determine whether sphinganine induces differentiation of type I HBEC to type H HBEC. Aim 5.8. Hypothesis. Sphinganine will induce differentiation of type I HBEC to type H HBEC to a similar degree as the known differentiating effect of sphingosine. Type II HBEC Aim 6. Determine effects of sphinganine on growth of type H HBEC in comparison to sphingosine. Aim 6. Hypothesis. Sphinganine will mimic sphingosine by having no or little effect on growth of type II HBEC. Transformed tumorigenic type I HBE C (M13S V1R2N1) Aim 7. Determine effects of sphinganine on growth and death of tumorigenic type I HBEC in comparison to sphingosine. Aim 7. Hypothesis. Sphinganine and sphingosine inhibit growth and cause death of tumorigenic type I HBEC. Aim 8. Determine whether sphinganine causes death of tumorigenic type I HBEC by inducing apoptosis in comparison to sphingosine. Aim 8 Hypothesis. Sphinganine and sphingosine will cause death of tumorigenic type I HBEC by inducing apoptosis. Aim 9. Determine whether sphinganine and sphingosine inhibit growth of tumorigenic type I HBEC by arresting the cell cycle at a specific phase. Aim 9 Hypothesis. Sphinganine and sphingosine will inhibit growth of tumorigenic type I HBEC by arresting the cell cycle at a specific phase. CHAPTER 2. LITERATURE REVIEW 10 A. Cancer A. 1. Cancer as a disease of proliferation and differentiation. A major characteristic of all higher eukaryotes is the defined life span of the organism. Normal cells appear to be "mortal", with highly regulated growth and division, and the potential to become terminally differentiated. Once cells are terminally differentiated, they can no longer proliferate (Trosko and Ruch, 1998). In contrast, cancer cells lose their usual growth control and proliferate indefinitely if provided adequate nutrients (Lewin, 1997) suggesting that cancer cells may be immortal (Kimball, 1983). Thus, a cancer can be described as an uncontrolled proliferation of cells. Cancer cells appear to originate from tissue precursor cells (stem cells), that are normally in a period of rapid mitosis and not yet fully differentiated. A common characteristic of cancer cells is that they are partially, but not terminally differentiated. Cancer cells are usually less differentiated than the cells of the tissue from which they arose. Thus, a malignancy also can be characterized by loss of differentiation called anaplasia (Markert, 1968) and cancer can be described as deregulation of differentiation or oncogeny as blocked or partially blocked ontogeny (Potter, 1978, Potter, 1987). Most cancers (~80%) originate fiom epithelial cells. Epithelial cells form the outer covering of tissues and surface cells of the skin and line the intestines and lungs. The surface cells are more likely to be exposed to dangerous chemicals and harmful radiation. The underlying layer must continue to divide as the outlayers are constantly worn away. Cells fi-om tissues, such as nerve and muscle, where cell division is rare, only occasionally become cancerous (Kimball, 1983, Lodish et al. 1999). 11 A.2. Cancer through mutations. Progression to cancer (carcinogenesis) occurs through multiple-stages including initiation, promotion, and progression phases. During the initiation phase, a single cell in a tissue acquires a mutation in an oncogene or tumor suppressor gene and stem cells are irreversibly altered. During the promotion stage, the initiated stem cells undergo clonal proliferation generating many descendants and these descendants are prone to further mutations in other genes that can lead to neoplasia. As the rate of mitosis in that clone increases, the chances of further DNA damage increase and many (perhaps 6-8) mutations may arise until the growth of that clone becomes completely unregulated. During the progression phase, cells within a tumor acquire further mutations that allow the angiogenic induction of vacuolization in the tumor. At this stage, cancer cells break away from the primary tumor, travel in blood and lymph, and establish metastases in other locations of the body. Metastases is normally a lethal stage of cancer in the patient (Kimball, 1983, Lodish et al. 1999, Trosko and Ruch, 1998). Mutations leading to cancer often occur in the following genes (Kimball, 1983, Lewin, 1997, Van Noorden et al. 1998, Lodish et al. 1999): 1) Cell cycle regulatory genes that regulate mitosis. These include proto- oncogenes (also called cellular genes) and tumor suppressor genes. Mutations or overexpression of prom-oncogenes (eg. erbB, the gene encoding the receptor for epidermal growth factor) stimulate mitosis although normal growth signals are absent. Oncogenes act as dominants meaning that only one of the pair needs to be mutated to predispose the cell to cancer. Tumor suppressor genes normally inhibit mitosis. For example, the p53 gene product normally senses DNA damage 12 and either halts the cell cycle until it can be repaired or, if the damage is too massive, triggers apoptosis. Tumor suppressor genes are recessive meaning that either both copies must be mutated for their function to be lost, or more commonly, the healthy copy of the gene has been lost. 2) Genes that regulate apoptosis (for details: see D.3. Induction of apoptosis by chemotherapeutic and/or chemopreventive agents). Mutations in these genes direct the cell to ignore signals telling it that it is irreparably damaged and should comnrit suicide. 3) Genes that maintain telomeres (for details: see D.5. Suppression of telomerase activity as a novel chemotherapy). 4) Genes that stimulate angiogenesis. Like other tissue, tumors require a blood supply to provide nutrients and oxygen and to remove waste. As a developing cancer grows, it must be able to stimulate the growth of new and normal blood vessels into itself. This is aided by the release of angiogenesis stimulants such as vascular endothelial growth factor. This may be the result of additional mutated oncogenes or tumor suppressor genes. 5) Metastasis genes'. These genes include: a) Genes whose products normally keep the cells of a tissue adhering to one another. For example, the E-cadherin genes help to hold epithelial cells together. b) Genes whose products normally keep the cells adhering to their extracellular substrate. For example, elevated expression of integrin genes encode proteases, which can break down the proteinaceous extracellular material that normally holds cells in place. 1 Genes that enable the tumor to separate from the primary tumor and migrate to other parts of the body 13 A.3. Environmental factors and cancer. Cancer is a disease which may be strongly influenced by environmental factors including diet, smoking, body mass, physical activity, and life style. Among many forms of cancers, colon and breast cancers appear to be particularly strongly affected by diet (World Cancer Research Fund and American Institute of Cancer Research, 1997). Thus colon and breast cancers might be preventable by appropriate diet, activity patterns, and reduction of other environmental exposures (World Cancer Research Fund and American Institute for Cancer Research, 1997) B. Colon cancer B. I . Colon cancer incidence. Colon cancer is the second leading cause of cancer mortality in the United States (American Cancer Society, 1996) and the fourth most common cause of cancer mortality in the world (World Cancer Research Fund and American Institute of Cancer Research, 1997). North America, Europe and Australia are areas of high incidence of colon cancer; whereas, Central and South America, Asia and Africa are areas of low incidence. Men and women have similar incidence rates of colon cancer (World Cancer Research Fund and American Institute of Cancer Research, 1997). 8.2. Colon cancer etiology and risk factors. 8.2.1. Colon carcinoma development. The adenoma-carcinoma hypothesis proposed by Hill et a1 (1978) is 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 14 phenotype. Bird et a1 (1987) suggested that hyperproliferation of the upper crypt cells leads to the formation of aberrant crypt foci (putative precancerous lesions of the colon) 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 (development of colon carcinoma) 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 (Figure 2.1) (V ogelstein et al. 1989, Fearon et al. 1990, Fearon and Vogelstein, 1990, Kinzler et al. 1991). The genes involved in colon carcinogenesis are: mutations or loss of the adenomatous polyposis coli (APC) gene (a tumor-suppressor gene) (Aaltonen et al. 1993), mutation of K-ras (a prom-oncogene), early disorganization of DNA methylation (Feinberg and Vogelstein, 1983), and late loss of p53 (a tumor- suppressor gene). 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 (El-Deiry et al. 1991). Several animal studies showed that isothiocyanates, which mainly are present in cruciferous vegetables, inhibited both carcinogenesis and DNA methylation (W attenberg, 1977, Wattenberg, 1987, Steinmetz and Potter, 1991a, & Steinmetz and Potter, 1991b). Although these observations are contradictory, both hypo- and hypermethylation of DNA are at least partially responsible for early stages of the carcinogenesis. 15 Normal epithelium Loss of APC (tumor suppressor gene) Abnormal cell division Hyperproliferation $4— DNA hypomethylation Adenoma 4— K-ras activation (oncogene) +— Loss of DCC (tumor suppressor gene) 4— Loss of p53 (tumor suppressor gene) Carcinoma ¢¢—— More mutations Metastasis Figure 2.1. Muti-step model for the colon carcinoma development (Adapted from Fearon and Volgetstein, 1990) 16 B.2.2. Family history as a risk factor for colon cancer. A family history of colon cancer is correlated with increased risk of colon cancer (Burt et a1. 1985, Bufill, 1990). One of the earliest studies of family history of colorectal cancer examined Utah families and reported a higher number of deaths from colorectal cancer (3.9%) among the first-degree relatives of patients who had died from colorectal cancer, compared to sex- and age-matched controls (1.2%) (Woolf, 195 8). Numerous studies which also have shown this difference have consistently established that first-degree relatives of affected cases are themselves at a two- to three-fold increased risk of colorectal cancer. The magnitude of colon cancer risk among family history groups is consistent in spite of the various study designs (case-control, cohort), sampling fiames, sample sizes, methods of data analysis, and countries where the studies originated (Duncan and Kyle, 1982, Rozen et al. 1987, St. John et al. 1993, Fuchs et al. 1994, Slattery and Kerber, 1994, Negri et al. 1998). 8.2.3. Genes associated with high risk for colon cancer. Several genes associated with colon cancer risk have been identified. All gene mutations recognized to predispose to colon cancer are inherited in an autosomal dominant fashion (Burt and Petersen, 1996). Thus, the family characteristics that implicate autosomal dominant inheritance of cancer predisposition are important indicators of high risk and of the possible occurrence of a cancer-predisposing mutation. For example, hereditary colorectal cancer has two forms: familial adenomatous polyposis (F AP, including an attenuated form of familial adenomatous polyposis, AP AP) and hereditary nonpolyposis colorectal cancer (HNPCC). 17 People with F AP possess gerrnline mutations in the APC gene and carry nearly 100 percent risk of colon adenocarcinoma (Aaltonen et al. 1993). HNPCC, in which the affected individuals have gerrnline mutations in DNA mismatch repair genes, is a syndrome which is not easily distinguished fiom 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). Other families displaying aggregation of colorectal cancer and/or adenomas, but without any apparent association with an identifiable hereditary syndrome are known collectively as familial colorectal cancer (FCC) (Burt and Petersen, 1996). B. 2. 4. Other risk factors for colon cancer. Putative risk factors for development of colon cancer include smoking (Potter et a1. 1993) and alcohol consumption (Potter et al. 1982, Tuyns et al. 1988, Longnecker, 1990, Choi and Kahyo, 1991). Other factors which appear to affect risk of colon cancer include level of physical activity (Garabrant et al. 1984, Gerhardsson et al. 1986, Slattery et al. 1988), age, and diet. Lee et a1 (1991) showed that individuals with high levels of physical activity were at lower risk for developing colon cancer and this observation partially accounted for the international differences in cancer rates (Garabrant et al. 1984, Gerhardsson et al. 1986, Slattery et al. 1988, Willett, 2001). 3.3. Dietary factors in colon caner. A large proportion of colon cancer deaths might be preventable by appropriate diets and behaviors (World Cancer Research Fund and American Institute of Cancer Research, 1997). In recent years, identification of specific beneficial food components which protect against chronic diseases such as colon cancer has been an area of focus in nutrition research. In this literature review, roles of 18 dietary fat and dairy and soy products on colon cancer are discussed since previously we found that bioactive compounds called sphingolipids are important components of dairy and soy products (Ahn and Schroeder, 2002a). B.3.1. Dietary fat. A hypothesis that a high-fat diet is responsible for colon, breast, and possibly prostate cancers in Western countries is rooted in the ecological correlations between national per capita fat consumption and rates of these major cancers (Armstrong and Doll, 1975, for review: Reddy 1993, Giovannucci and Goldin 1997, Lipkin et a1. 1999). This hypothesis also has been supported by animal studies (Cohen et al. 1986 Zevenbergen et a1. 1992, Tang et al. 1996, for review: Reddy 1993, Lipkin et al. 1999). It is noteworthy that both epidemiological studies and animal experiments have provided evidence that not only the amount of fat but also different types of fat play important roles in affecting colon cancer risk (for review: for review: Reddy 1993, Giovannucci and Goldin 1997, Lipkin et al. 1999). For example, saturated fat and/or animal fat has been shown to increase colon cancer risk, whereas fish products rich in w- 3 fatty acids, including docosahexanoic acid (DHA) and eicosapentanoic acid (EPA), has been associated with lowering colon cancer risk. Possible mechanisms of action of dietary fat and types of fat in colon carcinogenesis include: 1) diets high in beef tallow, lard, or corn oil increase the concentration of colonic luminal/fecal secondary bile acids, which promote colonic epithelial cell proliferation and function as promoters of colon cancer; 2) high dietary fat might alter membrane phospholipid turnover and prostaglandin synthesis by activating phospholipase PLA2, which enhances colon tumor promotion; and 3) w-3 fatty acids rich in fish oil have been shown to inhibit the cyclooxygenase (COX) pathway and 19 arachidonic acid (released fiom phospholipids) metabolism, which result in inhibiting prostaglandin synthesis. At least two isoforms of COX (COXl and COX2) are reported. Overexpression of COX2, but not COXl has been found in colonic mucosa and tumors of rats. On the other hand, the hypothesis of a high-fat diet as a risk factor for colon cancer has been argued with findings from prospective studies. For example, Slattery et a1 (1997) collected detailed dietary intake data flour a population of 1,993 colon cancer cases and 2,410 controls in 3 areas of the United States. Data indicated that neither total dietary fat nor specific fatty acids were associated with risk of colon cancer, after adjusting for total energy intake, physical activity, and body size (Slattery et al. 1997). In summary, ecological (correlational) studies and animal experiments support the association of a high-fat diet and colon cancer risk; whereas no association is seen in prospective cohort studies. A clear conclusion for dietary fat as a risk factor for colon cancer remains elusive. Dietary recommendations should not only consider total dietary fat intake but also types of fat, consumption of other nutrients and dietary components, and lifestyle factors that may influence colon cancer. 3.3.2. Caloric restriction. Energy or caloric restriction has gained much attention for its inhibitory effect on cancers. In addition, caloric restriction seems to be the only dietary treatment to improve longevity in laboratory animals. A case-control study indicated that increase of total calories is associated with risk of colon cancer (Graham et al, 1988). In rats treated with azoxymethane, caloric restriction reduced colon tumor growth (Pollard et al, 1984, Reddy et al, 1987). This cancer inhibitory effect appears to be independent of the effect of dietary fat because in chemically induced 20 models of colon and mammary cancers, 40% caloric-restricted rats showed reduced carcinogenesis even as they were fed increased percentage of dietary fat (Klurfeld et al, 1987) B.3.3. Dairy products. Studies have shown that dairy products appear to protect against colon carcinogenesis. Epidemiological studies indicate that regular consumption of fermented milk products such as yogurt may be protective against some forms of cancer (Peters et al. 1992, Kampman et al. 1994). In addition, 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-dimethylhydrazine-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. B.3.4. Soy producm. Several investigations suggest that soy products are another group of functional foods which contribute to the lower rates of colon cancer as well as breast and prostate cancers in China and Japan (Setchell et al. 1984, Barnes et a1. 1990, Adlercreutz, 1990). The isoflavone genistein, which is rich in soybeans and considered one of the most significant plant estrogens, has been reported to protect against development of colon carcinogenesis (Akiyama et al. 1987, Ogawara et al. 1989, Teraoka et a1. 1989, Thiagarajan et al. 1998). 21 C. Breast Cancer CI. Breast cancer incidence. Breast cancer is the most common cancer among women in the United States as well as among women worldwide and it is responsible for almost 30% of newly diagnosed cancers and 17% of cancer-related deaths (Parkin et al. 1992, Miller et al. 1993, Am. Inst. Cancer Res., 1997). Incidence rates are highest in North America and Northern Europe. Asia and Africa are the areas of the lowest incidence, although rates have been increasing markedly in several Asian and developing countries (Parkin et al. 1992). Among Caucasian women in the United States, age-adj usted breast cancer incidence rates have increased slowly for many decades, whereas age-adj usted breast cancer mortality rates have remained fairly constant since 1930 (Kelsy, 1993, Kelsey and Horn, 1993, Miller et al. 1993). The incidence of estrogen receptor-positive tumors has also risen over time (Glass and Hoover, 1990). The major factor accounting for the increase in incidence rates is improved breast cancer detection by screening of premenopausal women using mammography. Several factors might partially explain the increased incidence with no apparent increase in mortality: detection of early stage tumors by mammography; better prognosis associated with estrogen receptor-positive compared with estrogen receptor-negative tumors; and advances in medicine (Kelsy, 1993, Kelsey and Horn, 1993, Miller et al. 1993). C.2. Breast cancer etiology and risk factors. 612.]. Family history as a risk factor for breast cancer. Genetic contribution to breast cancer risk is found in the increased incidence of breast cancer among women 22 with a family history of breast cancer, and in the observation of rare families where multiple family members are affected with breast cancer, in a compatible pattern with autosomal dominant inheritance (genetic conditions that occur when a mutation is present in one copy of a given gene meaning that the person is heterozygous). Cross-sectional studies of adult populations reported that 5-10% of women have a mother or sister with breast cancer, and approximately 10-20% of women have either a first-degree or a second-degree relative with breast cancer. Taken together, epidemiological studies have clearly established that family history is an important risk factor for breast cancer. After gender and age, a positive family history is the strongest known predictive risk factor for breast cancer (American Cancer Society, 1999, Yang et al. 1998a, Colditz et al. 1993). C.2.2. Genes associated with high risk for breast cancer. Studies of large kindreds with multiple individuals affected by breast cancer led to the identification of several susceptibility genes including BRCAl, BRCA2, p53, and PTEN/MMACI. Women with gerrnline mutations in BRCAI or BRCA2 have a very high lifetime risk of developing breast cancer. Most of these germ-line mutations are predicted to generate a truncated protein product (For review: Sellers 1997). Furthermore, in all cancer studies from mutation carriers, the wild-type allele is deleted which strongly suggests that BRCA1 and BRCA2 belong to a class of tumor suppressor genes (Smith et al. 1992, Collins et al. 1995). The transcription of both BRCA1 and BRCA2 genes is induced late in the G1 phase of the cell cycle and remains elevated during the S phase, demonstrating a role in DNA synthesis (Gudas et al. 1996, Rajan et al. 1996, Vaughn et al. 1996). Interestingly, both BRCA1 and BRCA2 knockout 23 mice can be partially rescued by crossing with a p53 knockout strain, indicating that these genes may interact with the p5 3-mediated DNA damage checkpoint (Brugarolas and Jacks, 1997). Zhang et a1 (1998) provided further evidence that both BRCA1 and BRCA2 hereditary susceptibility genes function as " gate keepers" (similar to tumor suppressor gene p53) by serving to maintain genomic integrity. The loss of this function allows for the accumulation of other genetic defects that cause cancer. Breast cancer is a constituent of the rare Li-Fraumeni syndrome (LF S), where germline mutations of the p53 gene on chromosome 17p have been documented (Garber et al. 1991). The Li-Fraumeni syndrome is characterized by premenopausal breast cancer in association with childhood sarcoma, brain tumors, leukemia, and adrenocortical carcinoma (Bottomley et al. 1968, Malkin et al. 1993. Over 50% of families exhibiting this syndrome have shown a gerrnline mutation in the p53 gene. The germ line mutation of p53 is inherited with a penetrance of at least 50% by age 50 (for review: National Cancer Institute, 2002). It is known that individuals homozygous for ATM gene are at increased risk of malignancies, especially hematologic. Epidemiologic studies have suggested that female heterozygote carries of ATM gene might have an increased risk of breast cancer (Swift et al. 1987, Easton et al. 1994, Olsen et al. 2001). Ataxia telangiectasia is an autosomal recessive disorder described by neurologic deterioration, telangiectasias, immunodeficiency states, and hypersensitivity to ionizing radiation. By estimation, approximately 1% of the general population may be heterozygote carriers of the ATM gene (Savitsky et al. 1995). ATM proteins appear to play a role in cell cycle control (Uhrhammer et al. 1998) (for review: National Cancer Institute, 2002). 24 In summary, breast cancer occurs as a result of accumulation of mutations in critical genes that regulate cell proliferation, cell cycle, and differentiation. Most breast cancers appear as adenocarcinomas (a malignant tumor originating in epithelial cells in a glandular or gland-like pattern). Thus terminal end buds, which are rich in highly proliferating mammary epithelial stem cells, are considered to be the target cells of mammary neoplastic transformation (Russo and Russo, 1987). C.2.3. Other risk factors for breast cancer. Other risk factors that affect breast cancer among women include age, previous breast disease, reproductive and menstrual history, estrogen therapy, radiation exposure, alcohol intake (Davis et al. 1993), and diet (Marshall. et al. 1993). For example, cumulative risk of breast cancer increases with age and most breast cancers occur after the age of 50 (Feuer et al. 1993). In the case of women with a genetic susceptibility, breast cancer tends to arise at an earlier age than in sporadic cases. Breast cancer risk increases with early menarche and late menopause, whereas early first full term pregnancy decreases the risk. The Nurses' Health Study reported that these factors of reproductive and menstrual history influenced breast cancer risk only among women who did not have a mother or sister with breast cancer (Colditz et al. 1996) Conflicting data exist regarding the association between breast cancer and postmenopausal hormone replacement therapy. A meta-analysis of data from 51 studies showed a relative risk of breast cancer of 1.35 (95% CI 1.21-1.49) for women who had received hormone replacement therapy for 5 or more years afier menopause. The excess risk is reduced with termination of hormone replacement therapy and largely vanishes 25 within 5 years (Collaborative Group on Hormonal Factors in Breast Cancer, 1997). A recent study by Writing Group for the Women’s Health Initiative Investigators (2002) demonstrated that overall health risks exceeded benefits from use of combined estrogen plus progestin for an average 5.2 year follow-up among healthy postmenopausal US women. This study reported that women on hormone replacement therapy showed a higher rate of developing invasive breast cancer than the control group (Writing Group for the Women’s Health Initiative Investigators 2002). (3.3. Dietary factors in breast cancer. A large proportion of breast cancer deaths might be preventable by appropriate diets and behaviors (World Cancer Research Fund and American Institute of Cancer Research, 1997). In recent years, identification of specific beneficial food components which protect against chronic diseases such as breast cancer has been an area of focus in nutrition research. In this literature review, roles of dietary fat and dairy and soy products on breast cancer are discussed since previously we found that bioactive compounds called sphingolipids are important components of dairy and soy products (Ahn and Schroeder, 2002a). C.3.1. Dietary fat. The association between a high-fat diet and breast cancer has been an active area of research since early 1950. Tannenbaum and Silverstone (1953) demonstrated that a high-fat diet (12 %) stimulated mammary tumor development in mice compared to a low-fat diet (3 %). Ecological correlations exist between national per capita fat consumption and age-adj usted incidence and mortality from breast cancer (Armstrong and Doll, 1975; for review: Wynder et al. 1997, Willet 1997, Rogers, 1997, Lee and Lin 2000). Although correlational/ecological studies used food disappearance data, which do not reflect actual food-consumption habits due to differences in reporting 26 and wasted foods, the different fat intakes between countries have been confirmed by dietary surveys within national populations (for review: Wynder et al. 1997, Lee and Lin 2000). In addition, as Asian women with low fat diets increased their fat intake or migrated to western countries with higher fat diets, incidence of breast cancer increased (Osteen et al. 1986). Animal studies conducted over the past 50 years also support the hypothesis that a high-fat diet stimulates mammary turnorigenensis. A meta-analysis of 100 animal experiments showed an independent tumor-promoting effect of dietary fat (Freedman et al. 1990). Interestingly, several animal studies (Cohen et al. 1986, Zevenbergen et al. 1992, Tang et al. 1996) suggested a possibility that the tumor promoting effect of dietary fat intake may have a threshold level. For example, Tang et a1 (1996) examined effects of dietary fat ranging from 5 to 45% of calories, where fat was isocalorically substituted for carbohydrates. Data indicated that colon and mammary tumors rapidly increased when dietary fat level increased from 15 to 30 % of calories and no additional significant effects were seen on colon and mammary tumorigenesis above 30 % of calories fi'om fat (Tang et al. 1996). The mechanisms by which dietary fat promotes mammary tumorigenesis include: 1) conversion of essential fatty acid to eicosanoids; 2) generation of reactive oxygen species by reactions of oxygen and the conjugated double bonds in polyunsaturated fatty acids (PUFA); and 3) interaction between fatty acids and genomic DNA which alters gene expression (for review: Wynder et al. 1997, Lee and Lin 2000). Furthermore, it is important to note that different types of fat have different effects on breast cancer risk. Epidemiological studies showed that Mediterranean countries where olive oil is a staple showed lower breast cancer incidence compared to 27 most Western countries (for review: Esteve et al. 1993). For example, the incidence of breast cancer in Spain is about 40 % lower than that of North America or Northern Europe (for review: Esteve et al. 1993). Greek women consumed 42 % of calories from fat (mainly from olive oil) showed significantly lower rates of breast cancer than US. women who consumed 35 % of calories from fat (for review: Wynder et al. 1997, Lee and Lin 2000). Animal studies also indicated that high levels of olive oil and fish oil did not promote mammary tumorigenesis. However this concept of dietary fat as a risk factor for breast cancer based on correlational studies and animal experiments has been challenged. In a pooled anlysis of the six prospective studies with more than 200 cases of breast cancer (Hunter et al. 1996) did not find an association for total fat intake ranging < 20 to > 45 % of calories from fat with breast cancer. A large prospective cohort study of postmenopausal women (V elie et al. 2000) found little association between total fat (ranging < 28 to > 42 %) or fat subtypes and breast caner risk. A possible threshold effect of dietary fat observed in animal models (Cohen et al. 1986, Zevenbergen et al. 1992, Tang et al. 1996) may at least be partially related to this lack of association in cohort studies. In summary, correlational (ecological) studies and animal experiments support the association of a high-fat diet and breast cancer risk; whereas no association is observed in prospective cohort studies. A clear conclusion for dietary fat as a risk factor for breast cancer remains unsolved. Dietary recommendations should not only consider total dietary fat intake but also types of fat, consumption of other nutrients and dietary components, and lifestyle factors that may influence breast cancer. 28 C3. 2. Caloric restriction. Energy or caloric restriction has gained much attention for its inhibitory effect on cancers. In addition, caloric restriction seems to be the only dietary treatment to improve longevity in laboratory animals. Animal studies demonstrated that caloric restriction inhibits breast cancer incidence (Klurfeld et al. 1991). It is proposed that obese postmenopausal women have a higher risk of breast cancer (Cleary and Maihle, 1997) and overweight breast cancer patients have a poorer prognosis (Bastarrachea et al. 1994). 613.3. Dairy producm. Beneficial effects of fermented milk products are documented in epidemiological, animal, and cell culture studies. A case control study in the Netherlands showed a reduced risk of breast cancer among women who consume fermented milk products (Veer et al. 1989). Conjugated linoleic acid, butyric acid, ether lipids, and sphingomyelin found in milk fat components demonstrated anticarcinogenic effects in animal models (Parodi, 1997). Antiproliferative activity of fermented milk was demonstrated in MCF-7 breast cancer cells (Biffi et al. 1997). C.3.4. Soy products. Soy intake has gained much attention as an effective cancer fighter for years. Messina et a1 (1994) demonstrated that soy intake was inversely correlated with breast cancer risk among 142,857 women in Japan over a period of 17 years. Furthermore, studies of women in Singapore and China indicated that those who consume a traditional diet high in soy products had a low incidence of breast cancer (Lee et al. 1991, Wu et al. 1996). However, afier Asian women immigrated to the United States, the second generation (Ziegler et al. 1993), but not the first, loses this protection. These findings imply a possible mechanism for breast cancer protection fi'om early and specific nutritional exposure. Animal studies indicated that early exposure to estrogen 29 and progesterone caused differentiation of mammary tissue and reduced subsequent susceptibility to chemically induced mammary cancer (Russo et al. 1979, Grubbs et al. 1985). Genistein is an isoflavone found in soy and acts as a weak agonist to estrogen receptor. It also shows a property of an antioxidant, inhibits topoisomerase II and angiogenesis, and induces cell differentiation (Lamartiniere et al. 1997). Lamartiniere’s group proposes that breast cancer protection in Asian women consuming a traditional soy-containing diet is gained fiom early exposure to soybean products containing genistein (for review: Lamartiniere, 2000). D. Cancer chemotherapy and chemoprevention D.1. Chemotherapy. D. 1.1. Definition of chemotherapy. Chemotherapy is defined as the treatment of cancer with drugs that can destroy cancer cells by inhibiting their growth/proliferation. These drugs often are called “anticancer” drugs (also known as “anticarcinogens”). Side effects may occur when drugs affect healthy normal cells. Ofien two or more drugs are given at the same time because some drugs are more effective together than alone. This is referred to as combination chemotherapy (National Cancer Institute, 1999). D. 1.2. Goals for use of chemotherapy. Depending on the type of cancer and severity of cancer progression, chemotherapy is used for different goals: 1) Removal of cancer cells. When the patient remains free of evidence of cancer cells, cancer is considered cured. 30 2) Regulation of cancer cells. This is achieved by preventing cancer cells fiom spreading, by slowing growth, and by killing cancer cells. 3) Alleviation of symptoms caused by cancer. This can help patients live more comfortably (National Cancer Institute, 1999). Chemotherapy is often used in addition to surgery, radiation therapy, and/or biological therapy2 for the following reasons: 1) To shrink a tumor before surgery or radiation therapy. This is called neo-adjuvant therapy. 2) To help destroy any cancer cells that may remain after surgery and/or radiation therapy. This is called adjuvant chemotherapy. 3) To improve radiation therapy and biological therapy. 4) To help destroy cancer if it recurs or has spread to other parts of the body from the original tumor (National Cancer Institute, 1999). Some chemotherapy drugs might be used for just one or two types of cancer, while others are used for many different types of cancer. A chemotherapy treatment plan by a doctor is based on: 1) the type of cancer a patient has; 2) the location the cancer is found; 3) the effect of the cancer on normal body functions; and 4) the patient’s overall health (National Cancer Institute, 1999). D.2. Chemoprevention. Possible strategies to reduce the mortality and morbidity of cancer include: 1) prevention; 2) early diagnosis and intervention; 3) successful treatment of localized cancer; and 4) improved management of nonlocalized 2 Biological therapy, also called irnmunotherapy, is a treatment to stimulate or restore the ability of the immune system to fight against infection and disease. 31 cancer. Among these 4 strategies, the most practical approach for cancer management seems to be the prevention (Mukhtar and Ahmad, 1999). D.2. 1. Definition of chemoprevention. Chemoprevention can be defined as a means of cancer management in which the occurrence of the disease can be entirely prevented, slowed, or reversed by the administration of one or more naturally occurring and/or synthetic compounds (Ahmad et al. 1998). In addition, the definition of chemoprevention can be expanded as the use of agents (ofien called drugs) to prevent the occurrence of precancerous lesions or markers or to retard or reverse progression of premalignancy to malignancy (Kaityar and Mukjtar, 1996, Lippman et al. 1998). Chemoprevention is different from the treatment of established cancer. For example, when an established cancer is treated by chemotherapy, chemoprevention could prevent its recurrence and further development of new cancers. D. 2. 2. Characteristics of the chemopreventive agents. Ideal characteristics of the chemopreventive compounds (also known as anticancer drugs or anticarcinogens) for human usages are: 1) little or no adverse effects; 2) high efficacy against multiple sites; 3) effectiveness at achievable dose levels; 4) activity following oral consumption; 5) a known mechanism of action; 6) low cost; 7) history of use by the human population; and 8) general human acceptance (Mukhtar and Ahmad, 1999). D.2.3. Defense mechanism for damaged cells. Many environmental, dietary, and genetic factors impair the normal cell by different mechanisms at different targets. It is known that most humans have many initiated cells in the target tissues at all times (Jonason et al. 1996). These damaged cells, as a part of the defense mechanism, can react in different manners: 1) differentiate to other cell types without harboring the damages; 32 2) undergo apoptosis (programmed cell death) which eliminates the cell from the living organism; and 3) enter cell cycle arrest which prolongs the normal length of the cell cycle, thereby giving more time to repair damage. D. 3. Induction of apoptosis by chemotherapeutic and/or chemopreventive agents. D.3.1. Removal of unwanted cells via apoptosis. Apoptosis is a type of programmed cell death or cell suicide with distinctive morphological and biochemical changes that allows deletion of unwanted cells fiom an organism (V aux et al. 1996). Unlike tissue oncotic necrosis which occurs in response to severe insults and injury to cells, apoptosis involves an orderly breakdown of cells. In apoptosis, the nuclear chromatin is condensed and aggregates under the nuclear membrane. Then, activation of endonucleases causes fiagmentation of DNA into multiples of 200 base pair nucleosome- sized pieces. Cells shrink, exhibit cytoplasmic budding, and fragment into membrance- bound vesicles of cytosol and organelles called apoptotic bodies (Michaelson, 1991). Normally, apoptotic bodies are phagocytosed and degraded without eliciting an inflammatory response in the surrounding tissue (Walker et al. 1988). The presence of apoptosis can be determined by alterations in morphological appearance of nucleus and membrane and presence of endonucleosomal DNA fragments. The distinctive features of oncotic necrosis and apoptosis are represented in Table 2.1. 33 .aoéoo .oaaooooa dose .325: comanom .3 .& Aaoaoaooo ..m .3882 4 sass .d .5: ..m .5an ..> .838 ..< .oabé ooueoéea =3 o5 mafia... "a 38: 33:82 bassoon see 335 momcoamo. bofiEEmcg oz momeommoo Deng—a3 33:3..an .o 2.8 €08.65 3 33:68ch omfimosoa .3 mmm§8wfim =25? .mo_mo_o_w>ca >9 0339... 30595.68 Homwgommbaéo: .3 Back.“— o__oo _o=o_>__oc_ 66% Co 580 anon :8 co 689 .253 33233.... 33c 23% =8 ho Eo>o >53 Saginaw: <20 carbon. :8 .8 ago 8% Eggnog <29 25%.:— Amxmoan acute 03:3 conun- 520 .253: .825 <28 <75 mo aowmomao 8853 Gov F. 580 6: mean .3305 0288 23:33 ES; 335. “88.83% .358 oz 32» 23.555 new cozmzfim 9.295 3895 35:02 229... £33883 :3 mo 8332 mo 33 his =00 32w SE :3 3:2... ABEEEuoEV 352:. ABEESS 888 ifiaxm .258. 32.385 320:: 325.39.... 3226 USS ocmBEoE 952a 885 2.3g... engage mafia mo gamed USE EmEo. mo=mcw90 8:23qu mo 3:50.53 “"3385me $063 ozouaoamv wo_o_mo> canon 0555.2: 8 cosmetou 89.88 228., on £92 80385 $59.:th .33 533538 .2230 $9: can :8 05.8 mew—Ban 9.9589: 820:: 65 “a 5:32QO 55anch 52:85 .3 eouoaooofi 9.533 mews—:22 .0323 oqfinaoa mo no.3 non—soon 2.33—2.93: m_wo._.n_on_< m—momomz “Vb—bozo .mmmoaoqa 23 £8.80: mo magnum 8:35 ”88—6 :8 no 832 4." ~33. 34 D. 3. 2. Induction of apoptosis by chemotherapeutic agent. Data indicate that many successful chemotherapeutic agents selectively kill target tumor cells without influencing adjacent normal cells by inducing apoptosis (Sweeney et al. 1996, Hannun, 1997). Susceptibility to apoptotic induction varies by cell type suggesting that chemotherapeutic agents may induce apoptosis in tumor cells, while arresting the cell cycle in normal cell counterparts (David, 1994). Disruption of the apoptosis pathway in tumor cells may lead to a survival advantage or resistance to chemotherapeutic treatment (David, 1994). Alternatively, inappropriate stimulation of proliferation may cause apoptosis since prom-oncogenes including c-myc (Green, 1997), c-fos and c-jun (Preston et al. 1996), which stimulate cell proliferation, can also induce apoptosis. B.3.3. Induction ofapoptosis by chemopreventive agena. In recent years, apoptosis has gained considerable interest in biomedical research as the rate of apoptosis significantly affects the life span of both normal and cancer cells within a living system. Chemopreventive agents that can modulate apoptosis may be able to affect the steady- state cell population. Therefore, the chemopreventive agents, which have proven effects in animal tumor bioassay systems and/or human epidemiology and induce apoptosis in cancer cells, have great implications for cancer chemotherapy and chemoprevention. D. 4. Roles of stem cell ditferentiation for cancer prevention. D.4.I. Characteristics of stem cells. Stem cells are undifferentiated cells that are capable of proliferation and self-renewal, giveing rise to both differentiated descendants and more stem cells (Potten, 1997). Totipotent stem cells have unlimited capacity to specialize into extraembryonic membranes and tissues, the embryo, and all postembryonic tissues and organs. Pluripotent stem cells are capable of giving rise to 35 many types of cells but not all types of cells necessary for fetal development. (National Institute of Health, 2001, Trosko and Ruch, 1998). The relatively undifferentiated nature of tumor cells could be attributed to the blocked differentiation in stem cells or de- differentiation of differentiated cells that lead to cancer cells (V armur and Weinberg, 1993). As described in C. 2. Breast cancer etiology and risk factors, early full-term pregnancy decreases risk for breast cancer. This might be linked to stem cell multiplication that develops beginning at the time of puberty and during each ovarian cycle until the first pregnancy, but not after the first pregnancy (Cairns, 1975). Alternatively, pregnancy may induce full differentiation of the mammary gland (Russo et al. 1990), reduce the number of stem cells, and thus decrease the susceptibility for carcinogenesis. In a similar pattern, dietary components may decrease the risk of breast cancer by reducing the number of stem cells. D. 4.2. Differentiation genes. Although it is proposed that differentiation plays an important role in carcinogenesis (Prasad, 1991, Prasad et al. 1998), the specific genes that induce terminal differentiation in normal or transformed cells have not been identified yet. Prasad et a] (2001) emphasized the role of differentiation genes (genes that induce terminal differentiation in normal cells. Briefly, downregulation of a differentiation gene may be an initiating event to cause immortalization of normal cells. Such a gene can be referred to as a tumor- initiating gene in carcinogenesis. Secondary events may include mutations (point mutation or overexpression) in cellular oncogenes, growth regulatory genes, and tumor suppressor genes that may convert immortalized cells into transformed cells. Such genes 36 could be called tumor-promoting genes. Prasad et al’s differentiation gene hypothesis for carcinogenesis predicts the following concepts (for review: Prasad et al. 2001): “Tumor cells can be induced to differentiate terminally by appropriate agents if the differentiation gene is not mutated; Conversely, tumor cells with the mutated differentiation gene can be resistant to terminal differentiation”. 1) An elevation of 3’, 5’-cyclic adenosine monophosphate (cAMP) induces terminal differentiation in neuroblastoma cells despite the presence of extensive chromosomal anomalies and mutations in vitro and in vivo (Prasad, 1991, Prasad et al. 1994). This indicates that a differentiation gene is down-regulated due to alterations in the regulatory genes, rather than to a mutation in a differentiation gene, and thus a differentiation gene can be activated by an appropriate agent. 2) The existence of cAMP-resistant neuroblastoma cells suggests that such cells may carry a mutated differentiation gene. D.4.3. A mechanism of cAMP-induced differentiation. A possible mechanism of cAMP-induced differentiation is that an elevated cAMP level may activate a wild-type tumor suppressor gene (Li et al. 1998) or inactivate a differentiation suppressor gene. Hovland et al (2001) have identified some candidate “differentiation suppressor genes” that are down-regulated (N-myc, cyclin B 1, and protease nexin-I), and some candidate “differentiation genes” (c-fos, c-fes, and RAG-I gene activator) that are upregulated during cAMP-induced terminal differentiation in neuroblastoma cells (Hovland et al. 2001, for review: Prasad et al. 2001). D. 5. Suppression of telomerase activity as a novel chemotherapy. D.5. 1. Telomeres. Telomeres are repetitive TTAGGG sequences at eukaryotic chromosomal ends that are shortened by 50-200 base pairs after each cell division as a 37 result of the incomplete replication of the 5’ ends of DNA molecules (Shay et a1. 1993). Telomeres are needed for proper chromosome segregation during mitosis by preventing nuclease degradation and for end-to-end fusion of chromosomes during replication (Kirk et al. 1997). Telomere shortening afier each division is correlated with cell senescence in vitro and cell aging in vivo. The telomere hypothesis suggests that telomere length serves as a mitotic clock for timing cellular replicative life span (Autexier et al. 1996, Chin and Harley, 1997). D. 5.2. Telomerase. Telomerase is a specialized ribonucleoprotein polymerase that adds hexonucleotides (TTAGGG) onto human chromosomal ends thereby preventing the replication-dependent shortening of DNA. The human telomerase enzyme is composed of at least three components including a RNA (hTERC, human telomerase RNA component), a telomerase-associated protein (TEPl), and a telomerase catalytic subunit (hTERT, human telomerase reverse transcriptase) with sequence similarity to reverse transcriptase enzyme (for review: Prasad et al. 2001). D. 5.3. High telomerase activity in cancers. A high level of telomerase activity is readily detected in most tumor cells. For example, telomerase activity has been shown in 98% of bladder cancers (Kyo et al. 1997, Lin et al. 1996), 95% of colorectal cancers (Tahara et al. 1995a), 94% of neuroblastomas (Hiyama et al. 1995a), 93% of breast cancers (Hiyama et al. 1996), 85% of liver cancers (Tahara et al. 1995b), 84% of prostate cancers (Sommerfeld et al. 1996), 80% of lung cancers (Hiyama et al. 1995b), and 75% of oral carcinomas (Kannan et al. 1997). In contrast, the majority of studies have demonstrated that most adult normal human somatic cells show little or no telomerase activity. Several studies have shown 38 that telomerase activity is expressed in human germ line cells (testes and ovaries) (Wright et al. 1996), human embryonic stem cells (Thomson et al. 1998), human lymphocytes and hematopoietic progenitor cells (Hiyama et al. 1995, Norrback et al. 1996, Weng et al. 1996, Hohaus et al. 1997), candidate stem cells from the fetal liver (Yui et al. 1998), human epidermal cells expressing a basal cell marker (Y asumoto et al. 1996), and from the basal layer (Harle et al. 1996), and in human endothelial (Hsiao et al. 1997) and uroepithelial cells (Belair et al. 1997). In addition, Sun et al (1999) reported that normal human breast epithelial cells (HBEC) (type I and type H HBEC) have a low level of telomerase activity. Type H HBEC show even lower level of telomerase activity than type I HBEC with stem cell characteristics (Sun et al. 1999). The presence of high levels of telomerase in most tumor types and immortalized cells supported the hypothesis that increased telomerase activity is a critical event for continuous cell proliferation of immortalized cells (Bryan et al. 1997, Counter et al. 1994, Kim et al. 1994, Rhyu, 1995, Kyo et al. 1997, Lin et al. 1996, Tahara et al. 1995a, Hiyama et al. 1995a, Hiyama et al. 1996, Tahara et al. 1995b, Sommerfeld et al. 1996, Hiyama et al. 1995b, Kannan et al. 1997). Kiyono et a1 (1998) have proposed that both telomerase activity and inactivation of Rb/Pl 6 INK4a are required to immortalize human epithelial cells. D.5.4. Roles of telomerase activity on proliferation and differentiation. Belair et a1 (1997) have proposed that telomerase activity can serve as an indicator for levels of cell proliferation rather than transformation. High telomerase activity can also be used to classify the degrees of colorectal carcinomas (Okayasu et al. 1998), esophageal cancer (Asai et al. 1998), prostate cancer (Uemura et al. 1998) and leukemia (Zhang et al. 1996). 39 Moreover, Sun et a1 (1999) reported that human breast epithelial cells with stem cell features are more sensitive to telomerase activation and immortalization by SV40 large T antigen than those with a more differentiated phenotype. However activation of telomerase does not arise until middle [~60 cumulative population doubling level (cpdl)] or late (~100 cpdl) passages. This suggests that increased telomerase activity is acquired subsequent to immortalization. D. 5. 5. Suppression of telomerase activity as a novel chemotherapy. Researchers propose that suppression of telomerase activity may be a novel ‘adjuvant therapy’ (anticancer drugs or hormones given after surgery and/or radiation to help prevent the cancer from coming back) for the treatment of human cancer and detection of telomerase activity may be a significant tool to diagnose cancer (Schalken, 1998, Holt and Shay, 1999). Data suggest that telomerase activity is associated with aggressiveness of breast tumors and reflects anti-proliferative effects of chemotherapy. For example, Hooes et a1. (1998) reported that telomerase activity was detected in all breast cancer patients. A significant correlation was found between enzyme activity, and tumor size, lymph node status and stage. Telomerase activity appeared to increase in primary carcinomas with ongoing tumor progression, while all chemotherapy-treated tumors showed lower telomerase activity. D. 5. 6. Regulation of telomerase. The regulation of telomerase in cancer cells is not clearly understood. Ku et al (1997) demonstrated that a protein kinase C inhibitor, sphingosine inhibited telomerase activity in NFC-076 human nasopharyngeal cancer cells. In human breast cancer, telomerase activity is associated with cell cycle regulatory defects including overexpression of cyclin D1 and/or cyclin E (Landberg et al. 1997). 40 Absence of telomerase activity was seen in Gz/M-synchronized MCF-7 and MBA-435 breast cancer cells (Zhu et al. 1996). Kang et al (1999) reported that AKT (also called protein kinase B), which is known as anti-apoptotic, enhanced the telomerase activity in SK-MEL 28 human melanoma cells. Recently, Yang et a1 (Submitted) indicatated that sphingosine inhibited telomerase activity in transformed tumorigenic type I human breast epithelial cells (HBEC) suggesting a possible role of sphingolipids in regulation of telomerase in cancers. E. Sphingolipids as possible chemotherapeutic and chemopreventive agents E. 1. Sphingolipids structures. 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 called sphingosine (Figure 2.2). Sphingosine (trans-4-sphingenine) and sphinganine (without the 4,5-trans double bond) are the most prevalent free long-chain bases of most mammalian tissues, but the cellular concentration of sphinganine is usually lower than that of sphingosine (Menill, 1991). Sphingomyelin (N—acylsphingosine- l-phosphocholine or ceramide phosphocholine) is mainly composed of sphingosine as a sphingoid base backbone and contains an amide-linked fatty acid and a phosphorylcholine as a polar head group (Figure 2.2). Except for sphingosine l-phosphate, sphingoid bases, ceramides, sphingosylphosphorylcholine, and sphingomyelin, all other sphingolipids are designated glycosphingolipids since they contain carbohydrate head groups. 41 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). E. 2. Sphingolipid signaling path ways: Ceramide generation caused by sphingomyelinase activation. Ceramides and sphingoid bases (sphingosine and sphinganine) fimction in cells as second messengers in signal transduction pathways which regulate cell proliferation, apoptosis, and differentiation. Extracellular agonists including cytokines, growth factors, and hormones, stimulate their cell surface receptors to activate a sphingomyelinase which cleaves sphingomyelin to generate cellular ceramide (Figure 2.2.). For example, 10L,25-dihydroxyvitamin D3 (Okazaki et al. 1989), tumor necrosis factor-a (TNF-oc) (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), dexamethasone (Rarnachandran et al. 1990), complement components (N iculescu et al. 1993), and fimgal macrolide brefeldin A (Linardic et al. 1992) also may generate ceramide as a second messenger. Cz-ceramide and other short-chain water-soluble analogues of ceramide induced differentiation in HL-60 leukemia cells which mimicked the effects of 1a,25- dihydroxyvitamin D3, tumor necrosis factor alpha (TNF-a), and y-interferon on HL-60 cells (Okazaki et al. 1990). Treatment of U937 human myeloid leukemia cells with TNF- 42 0: caused sphingomyelin hydrolysis and resulted in elevation of level of ceramide. In addition, ceramide analogs or TNF-a caused intemucleosomal DNA fragmentation, a halhnark of apoptosis in myeloid and lymphoid cells (Obeid et al. 1993). 43 H WCHZOPO3'CH20H2N+(CH3)3 NH Sphingomyelin 3 NSAID H WHICH W" Ceramide 3 Ceramidase H MCHZOH Sphingosine "3+ Figure 2.2. Sphingolipids signaling pathway: Ceramide and sphingosine as mediators for protective effects of sphingomyelin against cancers. Abbreviations used are: 1,25-(OH)2 D3, 1a,25— dihydroxyvitamin D3; NSAID, nonsteroidal anti-inflammatory drugs. 1.2540le D: $ Sphingomyelinase Apoptosis E3. Sphingolipids occurrence and dietary sphingolipids. Sphingolipids are found in all eukaryotic and some prokaryotic cells and are located primarily in plasma membranes and related organelles including the Golgi apparatus, endosomes, and lysosomes which are functionally 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). For example, sphingomyelin is a phospholipid located mainly in the outer leaflet of the plasma membrane and accounts for 20% of plasma membrane lipid (Kolesnick et al. 1991) in most mammalian cells (Parodi, 1997). Furthermore, complex sphingolipids, as components of food, are found in dairy and soy products at relatively high concentrations (Vesper et al. 1999, Ahn and Schroeder, 2002a) E. 4. Prevention of colon carcinogenesis by dietary sphingomyelin. E. 4. 1. Uptake and metabolism of sphingolipids. Recently, sphingolipids have emerged as another component of the diet which may help to protect against development of colon carcinogenesis. Imaizumi et al (1992) reported that dietary sphingomyelin increased serum sphingomyelin in a concentration-dependent manner implying that dietary sphingolipids can also reach other organs via the circulation. Furthermore, using rodents, Schmelz et al (1994) showed that about 12% of dietary sphingolmyelin passes through the small intestine to the colon. These findings 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 2.2.). 45 15.4.2. Inhibition of colon carcinogenesis by sphingolipids in rodent models. The hypothesis that sphingomyelin metabolites ceramide and sphingoid bases could inhibit colon carcinogenesis 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 crypts in CF-l 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 (benign tumors) and lower percentage of adenocarcinomas (malignant tumors) (Schmelz et al. 1996). Also, the potential of synthetic sphingomyelins with saturated or unsaturated sphingoid base backbones to suppress the number of aberrant crypt foci was investigated using CF—l mice treated with 1,2-dimethylhydrazine (Schmelz et 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-palmitoylsphingomyelin) (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, efliciently reduced the number of aberrant colonic foci (Schmelz et al. 1997). Major milk glycosphingolipids including glucosylceramide, lactosylceramide, and ganglioside GD; effectively reduced aberrant colonic foci in CFl mice treated with colon 46 carcinogen 1,2-dimethylhydrazine (Schmelz et al. 2000). Therefore, the inhibitory effects of sphingolipids are not altered by the complexity of the sphingolipid head groups. In addition, diets supplemented with ceramide, sphingomyelin, glucosylceramide, lactosylceramide, and ganglioside GD3 reduced the number of tumors in all regions of the intestine in Multiple Intestinal Neoplasia (Min) mice, which have a mutation in the APC gene resulting in the expression of a truncated Adenomatous Polyposis Coli (APC) gene product (Schmelz et al. 2001). Therefore, the inhibitory effects of sphingolipids are not limited to the chemically induced colon cancer animal model. E. 5. Inhibition of human breast cancer cell growth by sphingolipids. Yang et a1 (Submitted) tested the potential of sphingolipids to serve as chemotherapeutic and chemopreventive agents for human breast cancer. The results indicated that both sphingosine and Cz-ceramiade (a short chain ceramide analog) inhibited proliferation and caused death of tumorigenic breast cells (in vitro neoplastically transformed type I human breast epithelial cells (HBEC) and MCF-7 human breast cancer cells) via apoptosis indicating that both sphingosine and ceramide can function as potential chemotherapeutic agents. Furthermore, concentrations of sphingosine cytotoxic to tumorigenic breast cells had no effect on normal type II HBEC. Sphingosine activated the tumor suppressor retinoblastoma protein by dephosphorylation and inhibited telomerase activity in transformed tumorigenic type I HBEC. Interestingly, sphingosine induced differentiation of type I HBEC stem cells to type H normal HBEC, which is more resistant to carcinogenesis; whereas, Cz-ceramide did not affect differentiation, suggesting that sphingosine might serve as a potential chemopreventive agent by reducing the number of target stem cells for neoplastic transformation. 47 E. 6. Ceramide and sphingoid bases as mediators for various anticarcinogenic agents. It has been reported that anticarcinogenic effects of various agents are associated with sphingolipid metabolism including the generation of ceramide via activation of sphingomyelinase (Figure 2.2.). For example, Chan et al. (1998) reported that sulindac, one of the most extensively investigated nonsteroidal antiinflarnmatory drugs (NSAH)), induced apoptosis in HCT-116 and SW-480 human colon cancer cells through increasing the level of arachidonic acid followed by stimulating conversion of sphingomyelin to ceramide. Furthermore, tamoxifen, widely used for breast cancer treatment, induced apoptosis in both estrogen receptor positive and negative human breast cancer cells (Frankfirrt et a1. 1995, Perry et a1. 1995, Gelmann et al. 1996, Mandlekar et al. 2000). The chemotherapeutic effect of tamoxifin is reduced when generation of acylated-sphingoid base is impaired, but is enhanced when the degradation of acylated-sphingoid base is blocked (for review: Senchenkov et al. 2001). B—sitosterol (the main phytosterol in the diet) inhibited HT-29 human colon cancer cell growth and caused a 50% reduction in membrane sphingomyelin suggesting that the conversion of sphingomyelin to simpler sphingolipids such as ceramide and sphingoid bases might be involved in the protective effects of B—sitosterol (Awad et a1. 1996, 1998). Also, Holtz et al. (1998) reported that B—sitosterol induced apoptosis in LNCaP human prostate cancer cells by increasing the concentration of ceramide. Cheng et al. ( 1999) indicated that ursodeoxycholic acid inhibits the development of rat colon carcinoma with increases in the activities of alkaline sphingomyelinase and caspase-3, a key regulatory protease in apoptosis. 48 Generation of ceramide was observed in photodynamic treatment mediated apoptosis of L5178Y mouse lymphoma cells (Separovic et al. 1997). Photodynamic therapy (PDT) is a novel cancer treatment which can destroy selectively malignant, premalignant, and benign lesions in patients and is initiated by a harmless photosensitizer. Perpheophorbide-a methyl ester (a photosensitizer) induced slow and sustained NF-kB activation by activating the acidic sphingomyelinase that released intracellular ceramide (Matroule et al. 1999). The tumor killing effect of ionizing radiation was enhanced by chelerythnine, a PKC inhibitor, via activating sphingomyelinase (Chmura et al. 1997), while acidic sphingomyelinase-deficient human lymphoblasts and mice are defective in radiation induced apoptosis (Santana et al. 1996). E. 7. Induction of apoptosis by ceramide and sphingoid bases. Although evidence is now emerging which suggests that dietary sphingolipids may protect against the development of colon cancer and possibly breast cancer, little is known about the mechanism. Protective effects of sphingolipids against some cancers may be the result of digestion of complex sphingolipids such as sphingomyelin to ceramide and sphingoid bases (sphingosine and sphinganine) which play important roles in regulation of cell growth, differentiation, and apoptosis. For example, ceramide induces apoptosis in JB6 tumor cells (Davis et al. 2000), while sphingosine and a methylated derivative, N,N- dimethyl sphingosine, induce apoptosis in a variety of human cancer cells including CMK-7, HL-60, U-937, HRT-18, MKN-74, and COLO-205 cells (Sweeney et al. 1996). E. 8. Regulation of apoptosis by Bcl-2 family genes. One possible protective mechanism of sphingolipids is that sphingolipids alter the expression of genes which modulate apoptosis. For example, Bcl-2 family genes (Reed, 1995) encode for 49 proteins which either can inhibit (e. g. Bcl-Z, BcI-xL) or promote (e. g. Bax, Bak) apoptosis (Shimizu et al. 2000). For example, in PC-12 cells ceramide induces apoptosis via the activation of caspase-3 and these effects were blocked by overexpression of bcI-2 suggesting that ceramide may activate caspase-3 by down-regulating bcl-2 (Yoshimura et al. 1998). 50 E. 9. Regulation of apoptosis by mitogen activated protein kinases MAPKs). E. 9.1. Major isoforms of MAPKs (ERK, JNK, and p38). Mammalian protein kinases are categorized into two major families that phosphorylate either serine and/or threonine residues (serine/threonine kinases) or tyrosine residues (tyrosine kinases). A major role for cellular kinases is to participate in signal transduction pathways through which cells respond functionally to external messages or to extracellular stresses. A family of serine/threonine mitogen-activated protein kinases (MAPKs) are activated by diverse stimuli (Cohen, 1997, Fanger et al. 1997, Ferrell, 1996, Marshall, 1995). Major isoforms of MAPKs include: Extracellular signal-regulated kinase—l (ERK1) and ERK2, also known as p44 and p42 MAP kinases, c-Jun NHz-terminal kinase/stress-activated protein kinase (JNK/SAPK), p38/RK/CSBP (p38), and big mitogen-activated protein kinase-l/ERKS (BMK-l/ERKS) (Cohen, 1997, Fanger et a1. 1997, Ferrell, 1996, Marshall, 1995, Kato et al. 1998) (Figure 2.3). [5.9.2. Roles of ERK, JNK, and p38. ERK] and ERK2 represent the prototypical mammalian MAPKs. ERK catalytic activation requires phosphorylation by MAP kinase kinase/ERK kinase kinase (MEK), exemplified by MEK-l (a dual specificity kinase), which in turn is phosphorylated and activated by a MAPK kinase kinase (also called MEK kinase) of the Raf family. Among these, c-Raf-l has been studied mostly and is activated at the plasma membrane after binding to the GTP-bound form of the low molecular weight G-protein p21ras (Cohen, 1997, Fanger et al. 1997, Ferrell, 1996, Marshall, 1995). As with ERK, JNK/SAPK, p38, and BMK-I/ERKS kinases are activated by phosphorylation that occurs at a specific threonine and tyrosine residue (Figure 2.3). 51 Substrate proteins that MAPKs phosphorylate include: additional ‘downstream’ serine/threonine kinases (which themselves become activated), cytoskeletal elements, cell death regulators, a number of nuclear receptors and transcription factors (for review: Camps et a1. 1999, Wilkinson and Millar, 2000). For example, ERK is linked to cellular chemotaxis, cell cycle progression, mitogenesis, oncogenic transformation, metastasis, neuronal differentiation and survival, and processes underlying memory and learning (Cohen, 1997, Fanger et a1. 1997, Ferrell, 1996, Marshall, 1995, Komhauser and Greenberg, 1997, Atkins et a1. 1998, Mansour et al. 1994, Cowley et al. 1994, Dudley et al. 1995, Reszka et al. 1997, Pages et al. 1993, Xia et a1. 1995). Emerging data suggest that the ERK pathway functions mainly to positively regulate the cell cycle dming G1 at the level of cyclin D synthesis, assembly of cyclin D-dependent kinase complexes and subsequent phosphorylation of retinoblastoma protein (Wilkinson and Millar, 2000). On the contrary, JNK/SAPK and p38 play important roles in pathways regulating T cell differentiation, production of inflammatory cytokines and eicosanoids, and apoptotic cell death (Cohen, 1997, Yang et al. 1997a, Yang et al. 1998b, Dong et a1. 1998, Xia et al. 1995, Saklatvala et al. 1996, Ichijo et al. 1997, Yang et al. l997b,Verheij et al. 1996, Zanke et a1. 1996). JNK/SAPK may oppose mitogenic functions of ERK and evoke a stress-induced cell cycle arrest. Accumulative studies have focused pathway identifications in immortalized cell lines using dominant negative or interfering mutants of JNK and p38. However, transformation of normal primary cells requires additional alterations in oncogenes or tumor suppressor genes (Figure 2.3). 15.9.3. Activations of ERK, JNK, and p38. Diverse cell stimuli preferentially activate distinct MAPKs. Growth factors, G-protein linked receptors, cell adhesion, 52 phorbol esters (known tumor promoters), and some oncogenes activate the ERKs, whereas inflammatory cytokines, trophic factor deprivation, and cell stresses preferentially activate JNK/SAPK and p38 (Cohen, 1997, Fanger et al. 1997, Ferrell, 1996, Marshall, 1995). Little is known about BMK-l/ERKS responsiveness, although Kato et a1 (1998) reported that both growth factors and stressful stimuli may activate the BMK-l/ERKS (Figure 2.3). E. 9. 4. Apoptosis triggered by imbalance between ERK, JNK, and p38. MAP kinases pathways do not function disj ointedly, instead they are integrated into networks of cellular signaling that affect upon and are influenced by MAP kinases. Furthermore, cells receive diverse stimuli that lead to integrated responses and cross-talk between multiple signaling pathways. Imbalance of these signals can alter transcription, cell cycle progression/arrest, and switch of cell survival versus apoptosis (Figure 2.3). E. 9. 5. Regulation of MAPKs by sphingolipids. Studies have indicated potential connections between sphingolipid signaling and MAP kinases pathways. For example, Cz-ceramide stimulated JNK activity and increased c-Jun mRNA levels, while C2- dihydroceramide had no effect in HL-60 human leukemia cells (W estwick et al. 1995). Jarvis et al (1997) showed that ceramide-mediated lethality is predominantly associated with strong stimulation of JNK2/JNK1 and weak inhibition of ERKl/BRKZ, whereas sphingosine-mediated lethality is largely associated with weak stimulation of JNK2/JNK1 and strong inhibition of ERKl/ERK2 in U937 human monoblastic leukemia cells. Sakakura et al (1997) reported that sphingosine and N,N-dirnethylsphingosine induced apoptosis and significantly inhibited ERK activity in tumor cell lines with high 53 ERK activity. In contrast, untransfonned cells and those tumor cell lines with low ERK activity displayed no significant change in activity and no apoptosis. Furthermore, high concentrations of Cz-ceramide, which induced apoptosis in the solid tumor cells, did not demonstrate significant effect on ERK activity. In an additional study, Sakakura et al. (1998) showed that D-erythro—sphingoinse was the most potent stimulator for ERK activity among D-erythro-sphingosine, L-threo-sphingosine, and DL-erythro- dihydrosphingosine tested, while DL-erythro-dihydrosphingosine was completely inactive. In summary, an altered balance between ERK, JNK, and p38 (Xia et al. 1995) may trigger induction of apoptosis by sphingosine and ceramide. 54 Growth factors (EGF, PDGF) Stress, UV, DNA damaging agent, Oxidant Mitogens, Hormones Osmotic and heat shock, Lipopolysaccharide Phorbol ester Inflammatory cytokines (T NFa, lL—1) ./ ‘s ,x \ if RTK / \ Ras / \ Raf (MKKK, MEKK) MEKK1,2,3 MEKK1,4 MKK1,2 (MEK1,2) MKK4,7(JNKK1,2) MKK3,6 l ERK1,2 JNK1,2,3 p38 Elk-1 1 1 Elk-1 Elk-1 cPLA2 ATF2 ATF2 MSK1 cJun cPLA2 PPARr MSK1 Proliferation, Apoptosis Figure 2.3. Mitogen Activated Protein Kinases signaling pathway. Abbreviations used are: EGF, epidermal growth factor; PDGF. platelet derived growth factor; TNFa, tumor necrosis factor-a; lL-1, interleukin-1; RTK, receptor tyrosine kinase; MKKK, MAP kinase kinase kinase; MKK, MAP kinase kinase; MEK, MAP/ERK kinase; MEKK, MAP/ERK kinase kinase; ERK. extracellular signal regulated kinase; JNK, c-Jun NH2-terminal kinase; JNKK, JNK kinase; cPLA2, cytosolic phopholipase A2; MSK1, mitogen-and stress- activated protein kinase-1; PPARr, peroxisome proliferator activated receptor gamma; CHOP. cAMP response element binding protein homologous protein; p90RSK, ribosomal p90 S6 kinase. 55 E.10. Regulation of apoptosis by AK T. E.10.1. AK T activation as cell survival and anti-apoptotic signal. AKT (also referred to as protein kinase B) is a serine/threonine kinase which plays a significant role in regulating the balance between cell survival and apoptosis (Zhou et al. 2000). An anti- apoptotic role of AKT is rooted fiom studies demonstrating that growth factors such as insulin, insulin-like growth factor-1, and nerve grth factor, which block apoptosis, increase activity of AKT by expressing either its constitutively active forms or its upstream regulators. Furthermore, enhanced AKT activity is associated with cancer progression in various tissues (Graff et al. 2000, Krasilnikov, 2000, Tsatasanis and Spandidos, 2000, Yuan et al. 2000). AKT appears to promote cell survival and to inhibit apoptosis by its ability to phosphorylate and inactivate several pro-apoptotic targets, including Bad (Cardon et al. 1998), caspase-9 (Rommel et al. 1999), and Forkhead transcription factors (Brunet et al. 1999, Kos et al., 1999). The cell survival and anti-apoptotic actions of AKT are presented in Figure 2.4. E] 0.2. Activation/phosphorylation of AK T at thr308 and serl 73. AKT has an amino-terminal pleckstrin homology (PH) domain that binds phosphorylated lipids at the membrane in response to activation of P13 kinases. AKT is activated via binding of phospholipids and phosphorylation of activation loop at thr308 by 3-phosphoinositide— dependent kinase (PDK) (Alessi et al. 1996) and also within the C-terminus at ser473 (Figure 2.4). E] 0.3. Regulation of AK T by sphingolipids. Recent studies evolve potential connections between sphingolipid signaling and AKT pathways. For example, a study in 56 Hep3B human hepatoma cell line by Chang et al. (2001b) demonstrated that sphingosine induced apoptosis and suppressed phosphorylated/active forms of AKT. Moreover, sphingosine-induced apoptosis was attenuated in cells transfected with constitutively active AKT. In HMNl motor neuron cells, Zhou et al. (1998) indicated that Cz-ceramide induced apoptosis and decreased both basal and insulin- or serum- stimulated AKT kinase activity. Furthermore, constitutively active AKT kinase reduced Cz-ceramide- induced cell death of HM] cells as well as COS-7 cells. Taken together, the AKT pathway is a probable target for actions of ceramide- (Zhou et al., Schubert et al, 2000) and sphingosine-induced (Chang et al, 2001b) apoptosis. 57 EGF, PDGF Insulin \, PI3K AKT l’\ Apaf-1 \ Iliad \ I 74 x ° " 14-3-3 , > ' , ,, z \ Caspase 9 Apoptosis (Death) Cell survival Figure 2.4. The cell survival and anti-apoptotic actions of AKT. Abbreviations used are: EGF, epidermal growth factor; PDGF, platelet derived growth factor; PI3K, phosphatidylinositol 3-kinase. Images in this dissertation are presented in color. 58 E.11. Involvements of Ca or calmodulin in proliferation and apoptosis pathways. Studies have shown that calcium and/or calmodulin play significant roles in DNA synthesis and cell proliferation (Chafouleas et al. 1982, Durha and Walton 1982, Parsons et al. 1983, Means 1988). There are conflicting data regarding the association between calcium channel blockers or calmodulin antagonists and cancer risk or cancer cell growth. For example, a cohort study by Sorensen et al. (2000) reported that there is no evidence of increased mortality of colon or breast cancer with administration of Ca channel blockers (verapamil, dihydropyridines, diltiazem, and mixed uses of these). Batra and Alenfall (1991) tested the effects of Ca antagonists (perhexilene, prenylamine, bepridil), Ca channel blockers (verapamil, diltiazem, nifedipine), calmodulin antagonists (pirnozide, R245 71, Trifluoroperazine), other compounds having a mixed Ca and calmodulin inhibitory effect on proliferation of HT-29 human colon cancer cells. The data suggested that in general, Ca antagonists had a moderate to strong inhibitory growth effects. Ca channel blockers were less effective and calmodulin antagonism seemed to be a common feature of compounds inhibiting cell proliferation in HT-29 cells. Recently Kim et a1 (2001) reported that fumonisin Bl, which blocks sphinganine (sphingosine) N-acyltransferase (also called ceramide synthase) and causes sphinganine accumulation, induced apoptosis by increasing the calmodulin expression at the levels of mRN A and protein. A calmodulin antagonist W7 attenuated fumonisin Bl-induced apoptosis in LLC-PKl pig renal epithelial cells (Kim et al. 2001). Different effects of W7 on apoptosis induced by various agents in a variety of cells are represented in Table 2.2. 59 «Bowman—me A8335 30 33=ooeb£v 3?. ammofiomee 632% so aa=8§é mo>ue>toe 28 0:30 M33 SHRED; .30: .5433 5 «oz .3 Ho anwexem Gaooém m=oo 820%? 5 8&5 28 0:30 Se a a $3: 830983 as Esta :22 a; ease :8 emf an; ongonfloho 8% Eomfiofieo/x mo> hm ooom .3 8 5 $30 3580: 323805 33889“ :0 “echo o5 02 mm _ Mme. 32 .3 3 3558M 8&8383— :8.» 333:8 33m £8 m oaoam oioonqfib Mae .3 a 3:3“ m 53 as am Ema €289. 3323 a.» 28 cream moo—533% £30 £332... .8 59 we 38.5— 93265 £332.44 2.8 we burg a S flame 33.3.» .3 @0265 $8239“ no $5 «a acute Bogota .N.N 03.; 60 E. 12. Summary: Mechanisms of actions of sphingolipids in proliferation, apoptosis, and differentiation. Cumulative data suggest that sphingolipids have potential as chemotherapeutic and chemopreventive agents based on their abilities to: 1) inhibit proliferation; 2) arrest cell cycle; 3) induce apOptosis in a variety of cancer cells; and 4) sphingolipids may induce differentiation of stem cells. Mechanisms by which sphingolipids inhibit colon and breast carcinogenesis are not clearly understood. Possible targets affected by sphingolipids to inhibit carcinogenesis include mitogen activated protein kinases (MAPKs), AKT, or Bel-2 family genes. 61 F. Cellular models to study chemotherapy and chemoprevention FJ. Human colon cancer cells. HT-29 and HCT-116 human colon cancer cells provide useful models to study cancer chemotherapy. Both HT-29 and HCT-l 16 cells possess adherent growth property and epithelial morphology and are tumorigenic in nude mice GBrattain et a1. 1981) (Figure 2.5-Photographs). HT-29 and HCT-l 16 cells show different mutation status in genes related to carcinogenesis and apoptosis (Table 2.3). F.1.I. H T-29 cells. HT-29 cells derived from colorectal adenocarcinoma harbor an APC tumpr suppressor gene mutation (Liu et al. 1997) and over-express a mutated nonfunctional p53 tumor suppressor gene (Rodrigues et a1. 1990, Redman et al. 1997), but carry normal B-catenin (Liu et al. 1997) and ras (K-ras, H-ras, N-ras) proto- oncogenes (Olivier et al. 1999). Also, HT-29 cells express c-myc, Myb, sis, fos oncogenes, and receptors for urokinase and vitamin D, but do not express N-myc oncogene (American Type Culture Collection, 2001). HT -29 cells express catalytically inactive cyclooxygenase 2 (COX2), which is also called prostaglandin H synthase 2 (PHSZ) (Hsi et al. 2000) (for review regarding COX2: Levy, 1997) (Figure 2.5, Table 2.3). F.1.2. HGT-116 cells. In contrast, HCT-116 cells derived from human colorectal carcinoma carry functional/normal APC (Groden et al. 1995, Narayan and Jaiswal, 1997), p53 (Zhang et al. 2000), box (pro-apoptotic gene) (Zhang et al. 2000). However HCT-116 cells have mutated B-catenin (Ilyas et al. 1997) and carry a mutation in codon 13 of the ras proto-oncogene and can be used as a positive control for 62 polymerase chain reaction (PCR) assays of mutation in this codon (Schroy et al. 1995). HCT-116 cells do not express COX2 (Kutchera et al. 1996, Hsi et al. 2000) (Table 2.3). F.1.3. H T -29 and HGT-116 cells for chemotherapy studies. HT-29 and HCT-116 human colon cancer cells can be good models to study chemotherapy as they show epithelial morphology and epithelial cells are the target cells of colon carcinogenesis (Hill et al. 1978) and are tumorigenic in viva. Furthermore, it is useful to study the gene targets of actions of sphingolipids as the two cells possess different mutation status in genes related to colon carcinogenesis including APC, ,B-catenin, ras, p53, and COX2. The differences in gene mutation status in HT-29 and HCT-116 cells are summarized in Table 2.3. 63 HT-29 . Her-116 -.'._ ’4 f' , ‘ ‘ “id" Human Colon Cancer Cells Type I Tumorigenic type I Type II Human Breast Epithelial Cells Figure 2.5. HT-29 and HGT-116 human colon cancer cells. Type I normal human breast epithelial cells (HBEC), tumorigenic type I HBEC, and type II HBEC (photographs). Images in this dissertation are presented in color. 050 33% 503822 me one .. 6353x0205 .X00 “:8 $8.58 2538983» .052 no.3 now: mucufi>2£< a2 a a 832.82.... .32 .3... «assuage 3.5:: e 252.. 88 a a a: 9.62. 2.8.538 .82 a a 8233. Basesaz 88 .3 a a... .3 58833 880 32 a a 3:3 33:2 82 .3 55.2.0 .382 we 32 a a 52.. uses: 52 .3 a a... 3582 555... 32 .3 a 5&8”. 88. a a 3:93 .aeoz .82 a .0 839.com 835.2 ma. 82 .353 Be €3.32 .32 a a 8.35 .aéoz 32 .a .o a: 835.2 92 nee—Shamem 253... 55352 «02.2301 «35» conga: 8:00 2.3 33b: 2.8 as... 38 6230 :28 38:: 2 Thu: 93 3.93 5 333 8358 8% E moonoeobfi ugh .QN 03:. 65 F.2. Human breast epithelial cells for chemoprevention study models. Human breast epithelial cells (HBEC) used in the current study were developed from tissues of healthy women obtained during reduction mammoplasty (Kao et al. 1995) (Type I and type H HBEC) (Figure 2.5; Figure 2.6; Figure 2.7; Table 2.4). Type I HBEC show stem cell characteristics and type H HBEC display basal epithelial cell phenotypes. In vitro neoplastically transformed type I HBEC (M13SV1R2N1) shows characteristics of tumorigenic breast cells (Figure 2.5; Figure 2.6; Figure 2.7). Derivation of type I HBEC, type II HBEC and neoplastically transformed type I HBEC are represented in Figure 2.7. Phenotypic differences between type I and type II HBEC are summarized in Table 2.4. F.2.1. T ype I HBEC as normal stem cells. Type I normal human breast epithelial cells (HBEC) display stem cell characteristics and are able to differentiate to type II HBEC (Figure 2.6), (Kao et al. 1995, Kang et al. 1997). Type I HBEC show variable shapes with smooth boundary, lack gap junctional intercellular communication, form budding/ductal structures on matri-gel (extracellular matrix), express estrogen receptor (Kang et a1. 1997) and luminal epithelial markers (e.g. epithelial membrane antigen, cytokeratin-IS, cytokeratin-19). Type I HBEC are more sensitive to telomerase activation and to immortalization after SV40 transformation (Kao et al. 1995, Sun et al. 1999). Therefore, type I HBEC appear to be the major target cells for breast carcinogenesis. Fetal bovine serum (FBS) stimulates type I HBEC growth. Type I HBEC and these simian virus 40 (SV 40) transformed cells can grow in an anchorage independent fashion (Kao et al. 1995). Phenotypic differences between type I and type II HBEC are summarized in Table 2.4. 66 F.2.2. Transformed tumorigenic type I HBE C. Derivation of in vitro neoplastically transformed HBEC lines fiom type I HBEC has been previously reported (Kao et al. 1995, Kang et a1 1998, for review: Chang et al. 2001a) (Figure 2.5; Figure 2.7). Type I HBEC are sequentially transformed to immortalized/non-tumorigenic cells (M13SV1), weakly tumorigenic cells (M13SV1R2), and highly tumorigenic cells (M138V1R2N1) by transfections with SV-40, x—ray, and neu oncogene (Figure 2.7). Transformed tumorigenic type I HBEC (M13SV1R2N1) are similar to breast carcinomas and possess many phenotypes of type I HBEC (eg. expression of epithelial membrane antigen, cytokeratin 18 and estrogen receptors, and deficiency in gap junctional intercellular communication) (Kao et al. 1995, Kang et al. 1997) (Table 2.4.). In the current study, transformed tumorigenic type I HBEC (MIBSVIRZNI) were used to represent a breast cancer cell model. F.2.3. Type II HBE C as normal cells. Type H HBEC do not express estrogen receptors. Type H HBEC express basal epithelial cell markers (e. g. a-6 integrin and cytokeratin-14), have gap junction genes (e.g. connexin 26 and 43), grow in an anchorage dependent fashion, and rarely become immortal afier SV40 transformation (Kao et al. I995, Sun et al. 1999). Fetal bovine serum inhibits type H HBEC growth (Table 2.4.). F.2.4. HBE C for chemotherapy and chemoprevention studies. Type I HBEC carry stem cell characteristics and are more susceptible to neoplastic formation indicating that type I HBEC are an excellent cell model for chemopreventive study since stem cells appear to be the targets of breast carcinogenesis. Chemotherapeutic potential of the 67 compounds can be studied in transformed type I HBEC as breast tumor cells in comparison to type H HBEC as normal breast cells. E2. 5. Criteria for identification of chemotherapeutic and chemopreventive agents in HBEC. The following criteria can be applied to identify the chemotherapeutic and chemopreventive properties of dietary compounds in Type I, transformed Type I, and Type H human breast epithelial cells: 1) Successful chemopreventive agents inhibit proliferation and induce apoptosis and differentiation in type I HBEC, but do not affect growth of type II HBEC. This will decrease the target stem cells for neoplastic transformation. 2) Successful chemotherapeutic agents inhibit proliferation and induce apoptosis in transformed type I HBEC. 68 Type II HBE Type I HBEC Type I HBEC After 24 h, After 2 weeks, mammary gland-like organoid ductal & terminal end bud-like structures Figure 2.6. Differentiation of type I HBEC with stem cell characteristics to type II HBEC and formation of ductal and terminal end bud-like structure from mixture of type I and type II HBEC (photographs). Type | HBEC are differentiated to type II HBEC over the course of time. Type I or a mixture of type I and type II HBEC have a unique ability to form mammary gland- like organoids after 24 h and ductal and terminal end bud-like structures after 2 weeks on Matrigel. Top panel: Magnification 10X plus 4X. The second and third photos are used with Dr. C.-C. Chang’s permission. Bottom panel: A photo on the left is obtained from Chang et al. 2001a and a photo on the right is from Sun et al. 1999, which are used with Dr. C.-C. Chang’s permission. Images in this dissertation are presented in color. Type I HBEC (stem cells) Type II HBEC Differentiation \ r t— SV40 transfection —-i , M13SV1_ lmmortalized at Immortallzed at '0“, fre uenc high frequency q y X-ray ‘\\\‘\\\-‘\“- ~\ \. \ V T \. a \__ M1 3SV1 R2 Weakly tumorigenic C-erbBZ/neu oncogene ‘T M13SV1R2N1 Highly tumorigenic Tumorigenic type I HBEC Figure 2.7. Derivation of tumorigenic type I HBEC and type II normal human breast epithelial cells (HBEC) from type I HBEC with stem cell characteristics. Abbreviation used: SV40, simian virus 40. 70 .388 .3 e 385 82. cameos. Aiéufioxoio v5. :585 0.8 .wov e33... :8 3.35...» 3am + + + 266:3 33 5.5.52”. 33 53935 £265 resonance omflonog 5380 2.3m mogosbm engage—o a. 3.5an 3215 pocfibmcgeoe a. 5095 we: genes mo 83% 0:33:58 .0923 E 8.53.23 8753.935 9.53335 .5035 0:35:58 3.65.3 dd. .33.: 3:953 3553 + 93.23 922 3:33: £952.28 093.. cos—358.3,. £390 £0533: 26:?28 __mE.wv 2.52335 $99.95 26.0 23.. .52. .2202.» 33:33:38.... E3569 3653. use EooEw ones» 5 032.3) 5.38 3.353 me. £5550 on 5.8980 .8932 cowobmm we muommmoaxm 5.32.5883 33:09.85 3:03.53. 9.0 835828 3 figummeom cougfito88_ €st .5233... owe—328 “coma...“ H ow...“— ov >m 8% 833.5833... came—mooz 888 0:33 Eva mo «Bum 538w “covaoaocfiowfionoé 333ng 5&3 333m came—q so Hangout... «@532 no 8.32:3 Easemeo awe—0:308 .8200 3293.58 :00 mam: : 2:9 0mm... _ a...» 6mm... 2.8 3.35...» .32.. 5&2. 38.8 a 25 38 . 25 =83». museums 0.2.82... .9." as; 71 CHAPTER 3 Sphingoid bases and ceramide induce apoptosu in [IT-29 and HCT-ll6 human colon cancer cells1 Running title: Sphingolipids and colon cancer cell apoptosis Key words: sphingosine; sphinganine; ceramide; colon cancer cells; apoptosis 1This chapter is published in Experimental Biology and Medicine (2002) 227:345-353 with authors Eun Hyun Ahn and Joseph J. Schroeder. 72 ABSTRACT Complex dietary sphingolipids such as sphingomyelin and glycosphingolipids have been reported to inhibit development of colon cancer. This protective role may be the result of turnover to bioactive metabolites including sphingoid bases (sphingosine and sphinganine) and ceramide which inhibit proliferation and stimulate apoptosis. The purpose of the present study was to investigate the effects of sphingoid bases and ceramides on growth, death, and cell cycle of HT-29 and HGT-116 human colon cancer cells. The importance of the 4,5-trans double bond present in both sphingosine and C2- ceramide (a short chain analog of ceramide) was evaluated by comparing the effects of these lipids to those of sphinganine and Cz-dihydroceramide (a short chain analog of dihydroceramide) which lack this structural feature. Sphingosine, sphinganine, and C2- ceramide inhibited growth and caused death of colon cancer cells in time- and concentration-dependent manners; whereas, Cz-dihydroceramide had no effect. These findings suggest that the 4,5-trans double bond is necessary for the inhibitory effects of Cz-ceramide, but not for sphingoid bases. Evaluation of cellular morphology via fluorescence microscopy and quantitation of fragmented low molecular weight DNA using the diphenylamine assay demonstrated that sphingoid bases and Cz-ceramide cause chromatin and nuclear condensation as well as fragmentation of DNA suggesting these lipids kill colon cancer cells by inducing apoptosis. Flow cytometric analyses confirmed that sphingoid bases and Cz-ceramide increased the number of cells in the A0 peak indicative of apoptosis and demonstrated that sphingoid bases and ceramide in HT29 cells and sphinganine and ceramide in HCT-116 cells arrest the cell cycle at Gle phase. These findings establish that sphingoid bases and ceramide induce apoptosis in colon 73 cancer cells and implicate them as potential mediators of the protective role of more complex dietary sphingolipids in colon carcinogenesis. 74 INTRODUCTION Recently, dietary sphingolipids have gained attention for their potential to protect against the development of colon cancer. Sphingolipids are a family of compounds which have a long-chain (sphingoid) base backbone and include free sphingoid bases (sphingosine and sphinganine), ceramides, sphingomyelins, cerebrosides, sulfatides, and gangliosides (Merrill et al. 1997). Several studies have been conducted with milk sphingomyelin, synthetic sphingomyelin, synthetic dihydrosphingomyelin, as well as milk glycosphingolipids to determine if the sphingolipids protect CFl mice from 1,2- dimethylhydrazine (DMID-induced colon cancer (Dllehay et al. 1994; Schmelz et al. 1996, 1997, 2000). The results showed that sphingomyelin at 0.1% of the diet caused a higher percentage of adenomas and lower percentage of the more advanced adenocarcinomas (Schmelz et al. 1996). Interestingly, synthetic dihydrosphingomyelin (N-palmitoyldihydrosphingomyelin) more potently reduced the number of aberrant colonic foci than synthetic sphingomyelin (N-palmitoylsphingomyelin) and milk sphingomyelin, suggesting that the 4,5-trans double bond, which is absent in dihydrosphingomyelin, is not required for the suppression of colon carcinogenesis (Schmelz et al. 1997). Furthermore, major milk glycosphingolipids including glucosylceramide, lactosylceramide, and ganglioside GD3 effectively reduced aberrant colonic foci in CF] mice treated with DMH (Schmelz et al. 2000). In addition, diets supplemented with ceramide, sphingomyelin, glucosylceramide, lactosylceramide, and ganglioside GD3 reduced the number of tumors in all regions of the intestine in Multiple Intestinal Neoplasia (Min) mice with a truncated Adenomatous Polyposis Coli (APC) gene product (Schmelz et al. 2001). Therefore, the inhibitory effects of sphingolipids are 75 not altered by the complexity of the sphingolipid head groups and are not limited to the chemically induced colon cancer animal model. Although evidence is now emerging which suggests that dietary sphingolipids may protect against the development of colon cancer, little is known about the mechanism. Sphingolipid metabolites such as ceramide and sphingoid bases play important roles in regulating cellular behavior and have been implicated as putative second messengers in signaling pathways (Merrill et al. 1997). For example, ceramide induces apoptosis in JB-6 tumor cells (Davis et al. 2000) and HT-29 human colon cancer cells (V eldman et al. 1998), while sphingosine and its methylated derivative N,N- dimethyl sphingosine induce apoptosis in a variety of human cancer cells including CMK—7, HL-60, U-937, HRT-18, MKN-74, and COLD-205 cells (Sweeney et al. 1996). Therefore, a possible mechanism by which complex dietary sphingolipids may reduce colon carcinogenesis is via digestion to bioactive metabolites such as ceramide and sphingoid bases. The purpose of the present study was to systematically examine the effects of sphingoid bases and ceramides on growth, apoptotic cell death, and cell cycle of PIT-29 and HGT-116 human colon cancer cells. Both cell lines show epithelial morphology, are tumorigenic in vivo, and are commonly used to study chemotherapy (Hill et al. 1978), but have different genetic mutations. HT-29 cells carry mutations in APC (Liu et al. 1997) and p53 (Rodrigues et al. 1990; Redman et al. 1997), but have normal ,B-catenin (Liu et al. 1997) and ms proto-oncogene (Olivier et al. 1999) and express catalytically inactive COX2 (His et al. 2000). In contrast, HCT-116 cells carry mutations in fl-catem'n (Ilyas et al. 1997) and ms (Schroy et al. 1995), but have normal APC (Groden et al. 1995) and p53 76 (Zhang et al. 2000) and do not express COX2 (Kutchera et al. 1996). In order to evaluate the significance of the 4,5-trans double bond, the effects of sphingosine and the cell permeable ceramide analog Cz-ceramide were compared to those of sphinganine and C2- dihydroceramide which lack this structural feature (Figure 3.1). Morphological and biochemical features of apoptosis were assessed via fluorescence microscopy, quantitation of fiagmented low molecular weight DNA using the diphenylamine assay, and flow cytometry. 77 OH WCHZOH NH3+ Sphingosine OH NH3+ Sphinganine OH WCHZOH \ 8 OH \ H3C\,NH C2-Ceramide 3 OH WCHZOH H3CJH Cz-dihydroceramide 3 Figure 3.1. Structures of sphingosine, sphinganine, ceramide, Cz-ceramide, and Cz-dihydroceramide. 78 MATERIALS AND METHODS Chemicals and reagents. Sphingolipids were obtained from Matreya (Pleasant Gap, PA). All other chemicals were from Sigma (St. Louis, MO) unless otherwise indicated. Sphingolipid treatment. Sphingosine and sphinganine were prepared as 1:1 complexes with bovine serum albumin (BSA) dissolved in phosphate buffered saline (PBS) while Cz-ceramide and Cz-dihydroceramide (cell permeable short chain analogs of naturally occurring ceramide and dihydroceramide, Figure 3.1) were dissolved in ethanol. Cell culture. HT-29 and HGT-116 human colon cancer cell lines were purchased from American Type Culture Collection (Rockville, MD). Stock cultures of HT-29 and HCT-116 cells were cultured in 100 mm dishes (Corning, Cambridge, MA) containing Dulbeco’s Modified Eagle Medium (DMEM) (Gibco BRL, Life Technologies, Gaithersburg, 1WD) supplemented with 10% fetal bovine serum (F BS) (GibcoBRL, Life Technologies, Grand Island, NY), 3.5 g glucose/L and 2.5 mL penicillin-streptomycin/L (GibcoBRL, Life Technologies, Grand Island, NY.) at 37°C and 5% C02. All experiments were performed using cells with from passage 20 or less. Assessment of cell proliferation. 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. Briefly, HT-29 or HGT-116 cells were seeded at a density of 3 x 105 cells/mL (i.e. 6.3 x 104/cm2 growth area) in 6-well dishes and cultured with 2 mL of DMEM with 10% FBS for 24 or 36 h to insure that cells were in log phase before treatment with sphingolipids. Then the medium was replaced with DMEM supplemented with 1% FBS. Various concentrations of sphingolipids were 79 added directly to each dish and the cells were cultured for up to 48 h. Following treatment, the medium and dead floating cells were removed, the viable attached cells were rinsed with 1 mL of phosphate-buffered saline (PBS), and then the cells were lysed with 1 mL of 0.1M NaOH. Total nucleic acid concentration was determined by measuring absorbance of the cell lysate at 260 nm using Gene Quant-RNA/DNA Calculator (Pharmacia Biotech, Piscataway, NJ). Fluorescence microscopic detection of apoptotic cells. To determine whether sphingolipids kill cells via apoptosis, cells were seeded at a density of 2 x 105 cells/mL (i.e. 4.2 x 104/cm2 growth area) in 6-well dishes and cultured with 2 mL of DMEM with 10% FBS for 24 h to insure that cells were in log phase before treatment with sphingolipids. Then the medium was replaced with DMEM supplemented with 1% FBS and cells were cultured with sphingolipids at 20 M for 24 h. Both attached and detached cells were collected, stained with acridine orange and ethidium bromide and examined via fluorescence microscopy (Mishell et al. 1980). Acridine orange intercalates into DNA and causes the DNA appear to green. It also binds to RNA but cannot intercalate so that the RNA stains red-orange. Ethidium bromide is only taken up by nonviable cells and intercalates into DNA, causing it to appear to orange, but binds only weakly to RNA. Thus, dead cells will have bright orange chromatin because the ethidium bromide overwhelms the acridine orange. Cells were photographed using a fluorescence microscope equipped with a camera under 10X plus 40X magnification with 400/490nm excitation and 520nm emission (Nikon Labophoto, Nikon Inc. Instrument Group, Garden City, NY.) Apoptotic and non-apoptotic cells were classified by the 80 differences in their chromatin organization as previously described (Martin and Lenardo, 1991) Isolation of fragmented low molecular weight DNA. Cells were seeded at a density of 2 x 105 cells/mL (i.e. 1.8 x 104/cm2 growth area) in 100 mm dishes and cultured with 5 mL of DMEM with 10% FBS for 24 h to insure that cells were in log phase before treatment. Then the medium was replaced with DMEM supplemented with 1% F BS and cells were cultured with sphingolipids at 50 W for 48 h. Afier treatments, both attached and detached cells were collected and pelleted via centrifugation (150xg, 5min) and the supernatant was removed. Lysis buffer at 200 uL was added and the cells were incubated for 10 min at 4°C. Afler centrifugation (14,000xg, 30 min, 4°C) the supernatant containing fiagmented DNA was transferred into a new 1.5 mL Eppendorf tube. Pellets were resuspended in 150 uL of TE buffer and were incubated with 2 uL of 40 mg/mL RNAase A at 37°C for l h and 4 uL of 40 mg/mL Proteinase K (Life Technologies-Gibco BRL, Rockville, MD) at 37°C for 1 h. Quantitation of fragmented low molecular weight DNA using the diphenylamine assay. Fragmented DNA was quantitated via the diphenylamine assay (Burton 1968). Briefly, 108 1.1L of 5 M perchloric acid was added to samples (final concentration of perchloric acid was 1 M) and samples were heated at 70°C for 15 min. Then diphenylamine reagent (1.5 g diphenylamine, 1.5 mL concentrated sulfiiric acid and 0.5 mL 1.6% aqueous acetaldehyde in 100 mL glacial acetic acid) at 1076 uL was added and the samples were incubated at 30°C for 15-18 h for color development. Salmon Sperm DNA (GibcoBRL, Grand Island, NY.) was used as a standard. Absorbance was 81 measured at 600 nm and the percentage of fragmented DNA was calculated as: (Fragmented DNA/Total DNA) x 100. F low cytometric analysis of cell cycle and population. Flow cytometric analysis was performed as described by Telford et al. (1994) with some modifications. Cells were seeded at a density of 2 x 105 cells/mL (i.e. 1.8 x 104/cm2 growth area) in 100 mm dishes and cultured with 5 mL of DMEM with 10% FBS for 24 h to insure that cells were in log phase before treatment. Then the medium was replaced with DMEM supplemented with 1% FBS and cells were cultured with sphingolipids at 35 M for 24 h. After treatments, both attached and detached cells were collected, trypsinized, and filtered through 40 um nylon sieve. Following centrifugation (150xg, 5 min), the supernatant was removed and PBS (pH 7.4) containing 50% FBS was added. Cells at 106 to 2x10‘5 cells/tube were prepared and fixed with 70% cold ethanol at a final concentration of 50-53%. After cells were stored at 4°C for 1-5 h, cells were pelleted by centrifugation (150xg, 5 min) and resuspended in 500 uL of PBS containing 4% FBS (Darzynkiewcz et al. 1980). Then, 1 mL DNA staining solution [4% FBS, 0.1% Triton X—100, 100 M EDTA, 0.05 mg/mL RNase A (50 units/mg), 50 ug/mL propidium iodide in PBS (pH 8.0)] was added and the cells were kept at 4°C for 1-2 h before reading using a FACS Vantage Flow Cytometer (Becton Dickinson, San Jose, CA). The total number of cells analyzed for each sample was 5000. The A0 peak represents apoptotic cells and is the hypo-diploid area (sub-Go/G 1) to the left of the Gn/Gl diploid peak. The percentage of cells in the A0 peak was estimated with the PCS Express version 1.0 software (De Novo Software, Thomhill, Ontario, Canada). The percentages of cells in Go/Gt, S, and szM phases were determined using 82 ‘Win cycle’, a Multi-cycle DNA content & cell cycle analysis software (Phoenix Flow Systems, Inc., San Diego, CA). The Win cycle software program does not include any cell populations outside of standard cell cycle populations when calculating areas, thus excluding sub-Go/G1 data. Statistical analyses. Data for total nucleic acids assay were analyzed by two-way factorial analysis of variance (AN OVA). After application of ANOVA, the significance of differences in the means between control and treatment groups at specific culture periods were evaluated by multiple comparisons using the Bonferroni method. Data for quantitation of fragmented DNA using the diphenylamine assay were analyzed by one-way AN OVA. Differences were considered significant at p<0.05. 83 RESULTS Sphingosine, sphinganine, and Cz-ceramide inhibit growth and cause death of H T-29 and HGT-116 human colon cancer cells. To determine the effects of sphingolipids on growth and death of human colon cancer cells, subconfluent HT-29 and HCT-116 cells were cultured with sphingosine, sphinganine, Cz-ceramide, or C2- dihydroceramide and then total nucleic acids were determined as an index of cell number (Li et al. 1990). For HT-29 cells, total nucleic acids in control cultures doubled over 36- 48 h (Figure 3.2). Sphingosine, sphinganine, and Cz-ceramide each caused concentration- and time-dependent decreases in proliferation of HT-29 cells. Specifically, addition of sphingosine (Figure 3.2) at 20 and 50 M significantly reduced total nucleic acid concentrations within 24 h by 47 and 62%, respectively (p<0.05) compared to the corresponding control. Sphinganine (Figure 3.2) at 10, 20, or 50 M significantly (p<0.05) reduced total nucleic acid concentrations within 24 h by 20, 28, and 80%, respectively compared to the corresponding control. Cz-ceramide (Figure 3.2) also caused a significant (p<0.05) reduction in the total nucleic acid concentrations at 20 and 50 M within 48 h. Culture with Cz-ceramide at 50 W for 48h completely killed all cells. In contrast to the sphingoid bases and Cz-ceramide, Cz-dihydroceramide had no effect on proliferation of HT-29 cells at concentrations as high as 50 M (Figure 3.2). Total nucleic acids in control cultures of HGT-116 cells doubled within 24 to 36 h, indicating slightly more rapid growth than for HT-29 cells (Figure 3.2). Similar to their effects on HT-29 cells, sphingosine, sphinganine, and C2-ceramide caused concentration- and time-dependent decreases in proliferation of HGT-116 cells. Specifically, addition of sphingosine (Figure 3.2) at 20 and 50 M significantly (p<0.05) 84 HT-29 m HCT-1 16 250 200 - 150 - 100 - o 150 r- 100 r Total Nucleic Acid (% of time zero) m _Dh-cer m _Dh-cer 150 150 100 100 50 50- o a L n a n o l A 4 n 1 012243648 012243648 Hours Figure 3.2. Effects sphingoid bases and ceramides on growth and death of HT-29 and HGT-116 human colon cancer cells. Subcontinent cells were cultured with sphingosine (SO), sphinganine (SA), Cz-ceramide (Cer), and Cz-dihydroceramide (Dh—cer) at O (a), 1 (O), 5 (V), 10 (V), 20 (Cl) and 50 (I) uM for 3, 12, 24, or 48 h. Total nucleic acids were measured as an index of cell number. Results are expressed as a percentage of the control value at the 0 h. Data are from two experiments and represent mean 1 SEM (n=8). Where an error bar is not shown, it lies within the dimensions of the symbol. Means at each culture period with an asterik (*) are significantly different (P < 0.05) from the corresponding control. 85 reduced total nucleic acid concentrations within 24 h by 65 and 68%, respectively compared to the corresponding control. Sphinganine (Figure 3.2) at 10, 20, or 50 M significantly (p<0.05) reduced total nucleic acid concentrations within 24 h by 46, 77, and 91%, respectively compared to the corresponding control. Cz-ceramide at 20 “M also caused a significant (p<0.05) reduction in total nucleic acid concentrations within 24 h by 36% and Cz-ceramide at 50 M killed all of the HCT-116 cells within 24 b (Figure 3.2). Unlike the sphingoid bases and Cz-ceramide, Cz-dihydroceramide did not inhibit proliferation at concentrations as high as 50 M (Figure 3.2). Fluorescence microscopic detection of apoptotic cells in H T -29 and HGT-116 cells. The proliferation studies demonstrated that the higher concentrations of sphingoid bases and Cz-ceramide not only inhibited proliferation, but also caused death of the colon cancer cells. To determine whether the sphingolipids kill the cells by inducing apoptosis, chromatin organization of HT-29 and HCT-116 cells was evaluated by staining with acridine orange and ethidium bromide using fluorescence microscopy (Mishell et al. 1980; Martin and Lenardo, 1991). Sphingosine, sphinganine, and Cz-ceramide at 20 M for 24 h each caused characteristic apoptotic morphological changes in both FIT-29 and HCT-116 cells (Figure 3.3). These included viable cells with apoptotic nuclei which contain bright green chromatin with highly condensed or fragmented structure as well as nonviable cells with apoptotic nuclei which contain orange chromatin with highly condensed or fragmented structure. The condensed and fragmented material is uniformly stained by the acridine orange and shows small bright circles with the overall cytoplasm not as bright as the cytoplasm of non-apoptotic control cells. In comparison, C2- 86 HT-29 HCT-1 16 Control SO SA Cer Dh-cer Figure 3.3. Effects of sphin oid bases and ceramides on chromatin and nuclear condensation in HT— 9 and HGT-116 human colon cancer cells (photographs). Subconfluent cells were cultured in the absence (control) or presence of sphingosine (SO), sphinganine (SA), Cz-eeramide (Cer). and Cz—dihydroceramide (Dh-cer) at 20 uM for 24 h. Cells were stained with acridine orange and ethidium bromide and photographed under fluorescence microscope. Bar=20 um. images in this dissertation are presented in color. 87 dihydroceramide had no effect and viable cells with normal nuclei contained bright green chromatin similar to control cells (Figure 3.3). Quantitation of fragmented low molecular weight DNA using the diphenylamine assay. To assess the percentage of cells that undergo apoptosis in response to sphingolipids, both attached and detached cells were collected 48 h afier treatment with each sphingolipid at 50 M and intemucleosomal fragmented low molecular weight DNA was quantitated using the diphenylamine assay (Burton 1968). For PIT-29 cells, control cultures had ~13% fragmented DNA (Figure 3.4). Sphingosine, sphinganine, and Cz-ceramide caused significantly greater DNA fragmentation at 21, 31, and 27%, respectively (p<0.05). In contrast, DNA fiagmentation in cells cultured with Cz-dihydroceramide was 17% which was comparable to controls (Figure 3.4). Results were similar for HCT-116 cells, although the control cultures had a higher level of DNA fragmentation at ~22% (Figure 3.4). Sphingosine, sphinganine, and C2- ceramide significantly increased DNA fragmentation to 37, 47, and 38%, respectively (p<0.05). In contrast, DNA fragmentation in cells cultured with Cz-dihydroceramide was 24% which was similar to controls (Figure 3.4). Flow cytometric analysis of cell cycle and apoptotic H T -29 and HGT-116 cells. The effects of sphingoid bases and ceramides at 35 M for 24 h on the populations of HT-29 and HCT-116 cells in each phase of the cell cycle were evaluated via flow cytometry (Telford et al. 1994; Darzynkiewicz et al. 1980). As shown in a representative histogram in Figure 3.5, sphingosine potently reduced the ntunber of HCT-116 cells with diploid DNA content (Go/G1 region) compared to control and caused a corresponding increase in the number of cells with hypo-diploid DNA content (sub-Go/Gl region) 88 HT-29 HCT-1 16 50 b 40 a 30 20 10 40 30. 20. 10.. 0 * I . Cont Cer Dh-ce oCont Cer Dh-cer u D I a: r N % DNA Fragmentation N O J O T Figure 3.4. Effects of sphingoid bases and ceramides on fragmented low molecular DNA in HT-29 and HOT-116 human colon cancer cells. Subconfluent cells were cultured in the absence (control, Cont) or presence of sphingosine (SO), sphinganine (SA), Cz-ceramide (Cer), and C,- dihydroceramide (Dh-cer) at 50 uM for 48h. Fragmented DNA were isolated and quantitated using the diphenylamine assay. Data are mean 1 SEM (n=3 for HT-29 cells; n=4 for HOT-116 cells). Means with different letters are significantly different (P <0.05). 89 100 " 3° " Control Sphingosine Cell Number o 160 320 480 640 800 DNA content Figure 3.5. Sphingosine increases the number of A0 (sub-GOIG1) cells indicative of apoptosis (one representative histogram). Subconfluent HCT- 116 cells were cultured in the absence (control) or presence of sphingosine at 35 uM for 24 h and stained with propidium iodide. The cell cycle and population were determined via flow cytometric analysis. 90 indicative of apoptosis (called A0). To quantify the degree of apoptosis, cells in the A0 peak were counted and expressed as a percentage of the total cell population. For HT-29 cells, only about 0.8% of the control cells were in the A0 apoptotic peak, while sphingosine, sphinganine, and Cz-ceramide increased the number of apoptotic cells to 2, 3, and 8%, respectively (Figure 3.6). In comparison, HT-29 cells cultured with C2- dihydroceramide were similar to control cultures having only ~1% in the A0 apoptotic peak. Similar but more profound effects were seen in HGT-116 cells. Control cultures had 1-5% of the cells in the A0 apoptotic peak, while sphingosine, sphinganine, and C2- ceramide increased the number of apoptotic cells to 39, 45, and 27%, respectively (Figure 3.6). In contrast, only ~2% of the cells cultured with Cz-dihydroceramide were apoptotic which was similar to control. The percentages of HT—29 cells in Go/Gl, S, and Gz/M (mean j; SEM) (Figure 3.7) for each treatment were: control for sphingoid bases (65.9 i 0.4; 25.6 i 1.5; 8.6 i 1.1), sphingosine (41.2 i 6.3; 45.7 i 4.7; 13.2 i 1.7), sphinganine (31.5 i 0.6; 55.8 i 0.8; 12.7 _+_ 0.3), control for ceramides (83.2 i 2.5; 11.0 i 1.5; 5.9 i 1.0), Cz-ceramide (40.5 i 6.2; 19.7 i 2.7; 39.9 i 3.5), and Cz-dihydroceramide (84.8 i 1.5; 9.4 i 1.4; 5.9 i 0.2). The percentages of HGT-116 cells in Go/Gl, S, and Gz/M (mean 1; SEM) (Figure 3.7) for each treatment were: control for sphingoid bases (72.3 j; 4.1; 16.3 :I; 2.3; 11.5 1; 1.8), sphingosine (48.8 i 2.3; 45.0 i 1.0; 6.2 1; 1.3), sphinganine (19.9 i 4.7; 36.3 i 5.7; 43.9 i 10.3), control for ceramides (73.0 i 1.0; 19.5 i 0.5; 7.6 i 0.4), Cz-ceramide (25.8 j; 1.6; 17.2 ft 0.6; 57.1 i 2.1), and Cz-dihydroceramide (69.0 j; 4.0; 23.0 i 1.0; 8.0 i 0.5). 91 HT-29 HOT-1 16 3.5. 50. 2.8L 4o. 2_1_ 30.. =-"_’1.4. 20. 80.7.. 13. .3 0 Cont 80 SA 3 10 30 g s. 2 o. 6. 18 f 4. 12 o\ 2. 6 0 0 Cont Cer Dh-cei Cont Cer Dh-cer Figure 3.6. Effects of sphingoid bases and ceramides on apoptotic cell number in HT-29 and HGT-116 human colon cancer cells. Subconfluent cells were cultured in the absence (control, Cont) or presence of sphingosine (SO), sphinganine (SA), Cz-ceramide (Cer). and C2- dihydroceramide (Dh-cer) at 35 111M for 24 h. DNA was then stained with propidium iodide, the cell cycle was examined via flow cytometric analysis, and the percentage of cells in the A0 (sub-Go/G1) region was estimated with the PCS express version 1.0 software. Data are mean 1 SEM (n=2). 92 HT-29 HGT-1 16 100 75 50 25 % Cells oCont Cer Dh-Cel 0 Cont Cer Dh-cer Figure 3.7. Effects of sphingoid bases and ceramides on cell cycle distribution of HT-29 and HOT-116 human colon cancer cells. Subconfluent cells were cultured in the absence (control, Cont) or presence of sphingosine (SO), sphinganine (SA), Cz-ceramide (Cer), and C,- dihydroceramide (Dh-cer) at 35 pM for 24 h. DNA was then stained with propidium iodide, the cell cycle was examined via flow cytometric analysis, and the percentage of cells in each stage of the cell cycle, including Go/G1 phase, //,; S phase, 0; and (3le phase, I, was determined using the Multi- cycle DNA content & cell cycle analysis software. The A0 (sub-6016,) cell population was not included in calculation of cell population. Data are mean 1 SEM (n=2). 93 Sphingoid bases and ceramide in HT-29 cells and sphinganine in HGT-116 cells caused a greater percentage of cells in the Gle and S phase compared to controls (Figure 3.7). This increase in S phase cell population was accompanied by decreased Go/Gl cells (Figure 3.7). Cz-ceramide caused accumulation of cells at G2/M with reductions in S and Go/G1 phases. These findings demonstrate that sphingoid bases and ceramide in HT-29 cells and sphinganine and Cz-ceramide arrested the cell cycle at the G2/M phase. 94 DISCUSSION Complex dietary sphingolipids including sphingomyelin and glycosphingolipids have gained attention for their potential to protect against the development of colon cancer. These compounds reduce the number of aberrant colonic foci and the percentage of adenocarcinomas in chemically initiated mice (Dllehay et al. 1994; Schmelz et al. 1996, 1997, 2000) and decrease the number of tumors in Multiple Intestinal Neoplasia (Min) mice (Schmelz et al. 2000). The mechanism by which sphingolipids inhibit colon cancer is not clear; however, it is known that suppression of colon carcinogenesis by complex sphingolipids is not dependent on the complexity. of the sphingolipid head groups (Schmelz et al. 2000, Schmelz et al. 2001). This raises the possibility that their bioactive digestion products ceramide and sphingoid bases may mediate the inhibitory effects of sphingolipids. These molecules are putative second messengers in cells and have been shown to reduce proliferation and induce apoptosis in a variety of cancer cells (Merrill et al. 1997; Davis et al. 2000; Veldrnan et al. 1998; Sweeney et al. 1996). This study demonstrates that both of the sphingoid bases sphingosine and sphinganine, as well as Cz-ceramide inhibit proliferation and cause death of both HT-29 and HGT-116 human colon cancer cell lines at 10-50 M. This concentration range of sphingoid bases and ceramide is similar to that previously shown to inhibit growth and/or cause differentiation or death in a variety of tumor cells (Davis et al. 2000; Veldman et al. 1998; Sweeney et al. 1996; Okazaki et al. 1990; Stevens et al. 1990a; Stevens et al. 1990b; Goldkorn et al. 1991; Endo et al. 1991; Obeid et al. 1993). The finding that a portion of the cell population survived the insult fiom sphingoid base and ceramide treatments and subsequently grew slowly between 24 and 48 h is probably due to metabolism of these 95 compounds either via incorporation into more complex sphingolipids or phosphorylation and degradation (Smith et al. 1995; Smith et al. 1997; Warden et al. 1999). Presently, it is not clear whether colonic tissue is normally exposed to the concentrations of sphingoid bases and/or ceramide used in this study. Schmelz et a1 (1994) reported that ~88% of dietary sphingomyelin is digested and absorbed as ceramides and sphingoid bases in the small intestine and small amounts of dietary sphingolipids appear in lymph (Nilsson 1968) and serum (Nilsson 1969). Therefore, colonic tissue could be exposed to ceramide and sphingoid bases via the circulation. Alternatively, ~12% of dietary sphingomyelin is not absorbed and passes directly into the colonic lumen (Schmelz et al. 1994) where it may be metabolized by colonic bacteria. This would provide a more direct route by which colonic tissues may be exposed to ceramide and sphingoid bases. Another important finding of this study is that Cz-dihydroceramide, which lacks the 4,5-rrans double bond present in Cz-ceramide, had no effect on proliferation or death of either HT-29 or HOT-116 human colon cancer cells. This result establishes that the 4,5-trans double bond is necessary for the growth inhibitory and cytotoxic properties of Cz-ceramide in human colon cancer cells, but is not required by sphingoid bases. This is consistent with the results of previous studies which showed that short-chain ceramides caused apoptosis in many systems, while dihydroceramides lacking the 4,5-trans double bond had no biological effects (Obeid et al. 1993; Tepper et al. 1995; Sawai et al. 1995; Brugg et al. 1996; Karasavvas et al. 1996; Xu et al. 1998). In contrast, both sphingosine and sphinganine inhibit growth and induce apoptosis in a variety of cell lines and tumor xenografis (Sweeney et al. 1996; Endo et al. 1991; Ohta et al. 1995; Jarvis et al. 1996). 96 Interestingly, previous studies have indicated that synthetic dihydrosphingomyelin (N- palmitoyldihydrosphingomyelin) which lacks the 4,5-trans double bond was even more effective than synthetic sphingomyelin (N-palmitoylsphingomyelin) at reducing the number of aberrant colonic foci in CF-l mice treated with DMH (Schmelz et al. 1997). Similarly in the present study, sphinganine (which lacks the 4,5-trans double bond) more potently increased DNA fragmentation than did sphingosine. Thus, complex sphingolipids may inhibit colon carcinogenesis via turnover to the free sphingoid bases (sphinganine and/or sphingosine) rather than to ceramide and dihydroceramide; however, fiom a practical perspective most complex sphingolipids present in food contain the 4,5- trans double bond (Ahn and Schroeder 2002). Jarvis et al. (1996) showed that a combination of ceramide and a sublethal concentration of sphingosine or sphinganine was more effective at causing death of two human myeloid leukemia cell lines HL-60 and U-937 than ceramide alone. Therefore, another way in which complex sphingolipids may inhibit colon carcinogenesis is via turnover to a mixture of ceramide, sphingosine, and sphinganine. The present study demonstrates that sphingosine, sphinganine, and C2-ceramide kill HT-29 and HOT-116 human colon cancer cells by inducing apoptosis. These findings are consistent with those of Veldman et al (1998) who showed that Cz-ceramide induces apoptosis in HT-29 cells. Our findings underscore the possibility of utilizing sphingoid bases as well as ceramide or synthetic analogs as chemopreventive or chemotherapeutic agents for human colon cancer. Many chemotherapeutic agents have been shown to kill susceptible cells via apoptosis. For example, nonsteroidal anti- inflammatory drugs (NSAID) such as aspirin (Qiao et al. 1998), sulindac (Piazza et al. 97 1997; Waddell et al. 1998), piroxicam (Waddell et al. 1998), and indomethacin (Erickson et al. 1999) inhibit colon carcinogenesis via induction of apoptosis. Also, it is noteworthy that the anticarcinogenic effects of many chemotherapeutic agents are associated with activation of sphingomyelinase and generation of ceramide (and possibly sphingosine). Chan et al (1998) reported that sulindac, the most extensively investigated NSAID, induced apoptosis in HOT-116 and SW-480 human colon cancer cells by increasing the level of arachidonic acid which stimulated conversion of sphingomyelin to ceramide. Chemotherapeutic agents which induce apoptosis appear to have the advantage of targeting individual cells without eliciting an inflammatory response in the surrounding normal tissue. Sphingosine may also have this property since it has previously been shown to induce ap0ptosis in neoplastically transformed HUVEC and rat mesangial cells (Sweeney et al. 1996) as well as human breast epithelial cells (Hong et al. unpublished data) but not in their primary culture counterparts. To our knowledge, this is the first study to demonstrate that sphingoid bases and ceramide in HT-29 cells and sphinganine in HGT-116 cells arrest the cell cycle at the G2/M phase and cause accumulation of cells in the S phase. Apoptosis triggered under these conditions is often found in response to suppression of DNA replication or DNA repair in drug-induced cell death and appears to involve inhibition of topoisomerase I or H (Del Bino et al. 1991). For example, DNA topoisomerase inhibitor I (camptothecin) or H (teniposide) induced apoptosis in HL-60 cells with S phase accumulation (Del Bino et al. 1991; Gorczyca et al. 1993). In addition, the anticancer drug 5-fluorouracil induced apoptosis by arresting the cell cycle at G2 phase in human breast cancer grated in nude mice (Okarnoto et al. 1996) and mitomycin C and etoposide induced apoptosis with G2/M 98 phase arrest and S phase accumulation in HGT-116 human colon cancer cells (Olivier et al. 1998). Folate deficiency-induced ap0ptosis was found to coincide with a block at 62M and accumulation of cells in the S phase (Huang et al. 1999). Also, diallyl disulfide isolated from garlic (which is believed to reduce tumor incidence and suppress tumorigenesis) arrested HCT-15 cells in the G2/M phase and increased the percentage of cells in the S phase (Knowles et al. 1998; Knowles et al. 2001). The mechanism by which sphingoid bases and ceramide induce apoptotic cell death is an area of active investigation, but is still not clearly understood. Though the present study did not examine targets of ceramide and sphingoid bases, there does not appear to be a role for p53 as a mediator of ceramide or sphingoid bases induced apoptosis in HT-29 cells because these cells over-express a mutated nonfunctional p5 3 (Rodrigues et al. 1990; Redman et al. 1997). Furthermore, Dbaibo et al. (1998) have shown that ceramide inhibits cell growth, arrests the cell cycle, and induces apoptosis, but does not increase p53 expression in HL-60 and U937 human leukemia cells lacking functional p53. In addition, Bax (a pro-apoptotic member of the Bcl-2 family genes) probably does not play a major role in ceramide-induced apoptosis in HT -29 and HCT- 116 cells. Though HT-29 cells do not appear to have mutations in Bax (Carethers and Pham 2000) and HGT-116 cells are heterozygous for Bax (Bax +/+, +/-, -/-) (Carethers and Pham 2000; Zhang et al. 2000), Kim et al (1999) found that Bax protein expression did not change in Cz-cerarnide-induced apoptosis of HT-29 cells and Zhang et al (2000) reported that Cz-ceramide-induced apoptosis was effected very little in HGT-116 cells carrying only Bax (-/-). 99 In summary, this study systematically evaluated the effects of the sphingoid bases, sphingosine and sphinganine, as well as Cz-cerarnide and Cz-dihydroceramide on growth, death, and the cell cycle of HT-29 and HGT-116 human colon cancer cells. The sphingoid bases and Cz-ceramide were found to kill colon cancer cells by inducing apoptosis; whereas, Cz-dihydroceramide was not effective (Figure 3.8). These data suggest that ceramide and the sphingoid bases have the potential to mediate the protective effects of more complex dietary sphingolipids and raise the possibility that they themselves may be effective chemopreventive and chemotherapeutic agents for human colon cancer. Sphingoid bases appear to specifically arrest the cell cycle at G2/M and cause accumulation of cells in the S phase similar to apoptosis induced by several other anticancer drugs. 100 Colon cancer cells \ - - ~L Proliferation Sp hlngosme T Apoptosis 8: Arrest at GZIM . . Jr Proliferation Sphinganine TT Apoptosis 8. Arrest at 6le Ceramide ~L Proliferation T Apoptosis & Arrest at 62m Dihyd roceramide No effect J Figure 3.8. Regulation of cell growth, cell cycle, and apoptosis by sphingoid bases and ceramides in HT-29 and HOT-116 human colon cancer cells. 101 Acknowledgements We would like to express our thanks to Dr. Louis King and Dr. Zahidul Islam for the technical advice on flow cytometry analysis and to Dr. Ludmila Roze and Rebecca Uzarski for advice on fluorescence microscopic detection of apoptotic cells. We also express our appreciation to Dr. James Pestka and Dr. John Linz for providing access to their fluorescence microscope. 102 CHAPTER 4 Sphinganine causes early activation of JNK and p38 mitogen activated protein kinase andearly inhibition of AKT in HT -29 human colon cancer cells1 Running title: Regulation of ERK, JNK, p3 8, and AKT by sphinganine Key words: sphinganine; ERK; JNK; p38; AKT; apoptosis 1This chapter is prepared for publication with authors Eun Hyun Ahn and Joseph J. Schroeder. 103 ABSTRACT The sphingoid base sphinganine inhibits growth and induces apoptosis in HT—29 human colon cancer cells more potently than other bioactive sphingolipid metabolites sphingosine and Cz-ceramide tested in our previous study (Ahn and Schroeder, 2002b). The objectives of the current study were to investigate the effects of sphinganine at a concentration-inducing apoptosis on the mitogen activated protein kinases (MAPKs) and AKT (protein kinase B), which regulate cell proliferation and apoptosis. Major isoforms of MAPKs include extracellular signal-regulated kinase (ERKl/ERKZ), c-Jun NH;- terminal kinase (JNK2/JNK1, also called stress-activated protein kinase (SAPK)), and p38 mitogen activated protein kinase. HT-29 cells were cultured with sphinganine at 35 M and the protein expression and/or phosphorylation status of ERKI/ERKZ (p44/p42), JNK2/JNK1 (p54/p46), and p38 mitogen activated protein kinase, AKT-ser473, and AKT-thr308 were determined using Western blot analysis. Sphinganine had little or no effect on the protein expression level of any of the kinases. In contrast, sphinganine clearly increased the active phosphorylated forms of JNK2/JNK1, and p38 mitogen activated protein kinase at 15, 30, and 60 min, with minimal effects on activation of ERKllERK2. Sphinganine inhibited phosphorylation of AKT at ser473 at 60 min and thr308 at 15 and 60 min. The findings are consistent with a mechanism by which sphinganine induces apoptosis in HT-29 human colon cancer cells via early inhibition of AKT and early activation of JNK and p38 mitogen activated protein kinase. 104 INTRODUCTION Sphingolipids are dietary constituents (Ahn and Schroeder, 2002a) that have gained much attention for their potential to protect against the development of colon cancer (for review: Vesper et al. 1999). Complex dietary sphingolipids including sphingomyelin, dihydrosphingomyelin, glucosylceramide, lactosylceramide, and ganglioside GD3 reduced aberrant colonic foci in CF 1 mice treated with DMH (Schmelz et al. 1996, 1997, 1998, 2000) and the number of tumors in all regions of the intestine in Multiple Intestinal Neoplasia (Min) mice with a truncated Adenomatous Polyposis Coli (APC) gene product (Schmelz et al. 2001). This protective role of sphingolipids against colon carcinogenesis may be the result of conversion of complex sphingolipids to bioactive metabolites including sphingoid bases (sphingosine and sphinganine) and ceramide, which inhibit proliferation and induce apoptosis (programmed cell death) in various cancer cells (Sweeney et al. 1996, Ahn and Schroeder, 2002b). For example, sphingoid bases and ceramide inhibit growth and arrest the cell cycle at 02M phase in HT-29 and HGT-116 human colon cancer cells (Ahn and Schroeder, 2002b). Furthermore sphingoid bases and ceramide cause chromatin/nuclear condensation and DNA fiagmentation and increase the number of cells in the sub-Go/Gl phase, which are indicative of apoptosis, with sphinganine being the most potent among Cz-ceramide and sphingoid bases tested (Ahn and Schroeder, 2002b) A possible mechanism by which sphinganine could inhibit proliferation and induce apoptosis is by altering the balance of anti-apoptotic and pro-apoptotic signaling through mitogen activated protein kinases (MAPKs) and AKT. MAPKs are a family of 105 serine/threonine kinases and major isoforms include: extracellular signal-regulated kinase (ERKl/ERK2, p44/p42), c-Jun NHz-terminal kinase (JNK2/JNK1, p54/p46, also called stress-activated protein kinase (SAPK)), and p38 mitogen activated protein kinase (Cohen, 1997, Fanger et a1. 1997, Ferrell, 1996, Marshall, 1995, Kato et al. 1998). JNK and p38 mitogen activated protein kinase appear to have pro-apoptotic roles. Although ERK activation has most often been thought of as anti-apoptotic and pro-mitogenic, recent studies showed that ERK activation is not always required for mitogenic (Tharnilselvan et al. 2002) and anti-apoptotic (Chang et al, 2001b) effects. Sphingoid bases could induce apoptosis by altering the balance between ERK, JNK, and p38 mitogen activated protein kinase activation. For example, Jarvis et al (1997) reported that sphingosine-mediated lethality is largely associated with weak stimulation of JNKZ/JNKI, strong inhibition of ERKl/ERKZ, and enhanced p38 mitogen activated protein kinase activity in U937 human monoblastic leukemia cells. Alternatively, sphingosine could induce apoptosis by inhibiting AKT (protein kinase B). AKT is a cytoplasmic serine/threonine kinase that promotes cell survival and inhibits apoptosis (for review: Testa and Bellacosa, 2001, Talapatra and Thompson, 2001). Enhanced AKT activity is associated with cancer progression in various tissues (Graff et al. 2000, Krasilnikov, 2000, Tsatasanis and Spandidos, 2000, Yuan et al. 2000, Sun et al. 2001). Chang et al (2001b) demonstrated that sphingosine induced apoptosis and suppressed phosphorylated/active forms of AKT. Moreover, apoptosis induced by sphingosine was attenuated in cells transfected with constitutively active AKT. The hypothesis of the present study is that altered activation of MAPKs and AKT may regulate inhibition of proliferation and induction of apoptosis by sphinganine 106 observed in HT-29 human colon cancer cells. The influence of a concentration-inducing apoptosis of sphinganine was examined for effects on the protein expression and/or phosphorylation of MAPKs (ERKl/ERKZ, JNK2/JNK1, p38) and AKT (AKT-ser473 and AKT-thr308). 107 MATERIALS AND METHODS Sphinganine preparation. Sphinganine (D-erythro—dihydro-sphingosine) was obtained from Matreya Co. (cat. no. 1831, State College, PA). Stock solutions of sphinganine were prepared by dissolving sphinganine in ethanol at 50 mM and storing at 4°C. A working solution of sphinganine was prepared as a 1:1 complex with 1 mM bovine serum albumin (BSA) dissolved in phosphate buffered saline (PBS) and stored at 4°C up to one month. Prior to use of sphinganine for experiments, sphinganine was incubated at 37°C overnight and sonicated for 2 h to ensure sphinganine-BSA complex formation. Cell culture. HT-29 human colon cancer cells were purchased from American Type Culture Collection (Rockville, MD). HT-29 cells were cultured in 100 mm dishes (Corning, Cambridge, MA) containing Dulbeco’s Modified Eagle Medium (DMEM) (Invitrogen Life Technologies, Carlsbad, CA) supplemented with 10% fetal bovine serum (FBS) (Invitrogen Life Technologies, Carlsbad, CA), 3.5 g glucose/L and 2.5 mL penicillin-streptomycin/L (Invitrogen Life Technologies, Carlsbad, CA) at 37°C supplied with 5% C02 and humidified air. All experiments were performed with cell passage $20. Isolation and quantitation of cellular protein. Confluent HT-29 cells were trypsinizedl and seeded at 2 x 105 cells/mL (i.e. 1.8 x 104/cm2 growth area) in 100 mm dish and cultured in 5 mL of DMEM supplemented with 10 % FBS and 3.5 g glucose/L and 2.5 mL penicillin-streptomycin/L for 24 h to insure that cells were in log phase before treatment with sphinganine. Then the medium was replaced with DMEM supplemented with l % FBS, 3.5 g glucose/L, and 2.5 mL penicillin-streptomycin/L and sphinganine was added directly to each dish for various culture periods. Cells on ice 108 were extracted with lysis buffer (1.5 M Tris-Cl, pH 8.8, 100mM sodium vanadate, 20% SDS) and cell lysates were sonicated for 5-10 sec with a probe sonicator and stored at - 80°C until use. The cellular protein was quantitated via the Bradford assay (Bio-Rad Laborotories, Hercules, CA) (Bradford 1976). Western blot analysis for protein expression of ERKI/ERKZ, JNKZ/JNKI, and p38, and phosphorylated forms of ERKI/ERKZ, JNKZ/flVKI, p38, AK T-ser4 73, and AK T -tlrr308. Protein (20 pg for both phospho-ERK, JNK, p38 and ERK, JNK, p38; 60 pg for phospho-AKT-ser473; 40 pg for phospho-AKT-thr308) was applied and separated on 10% SDS-PAGE gel under reducing conditions and transferred to PVDF membranes at 80 V for 20 min and 120 V for 1.5 h. The blots were washed three times and blocked with Tris buffered solution-T (TBST, 100mM Tris-Cl pH 7.5, 0.9% (=150mM) NaCl, 0.1% Tween-20) containing 5% BSA for 8 h at 4°C and washed twice with TBST. The blots were incubated with primary antibodies (Cell signaling/New England Biolabs, Beverly, MA), (1:10,000 for anti-rabbit IgG polyclonal antibodies against phospho- ERKl/ERKZ, phospho-JNKZ/JNKI, phospho-p38, ERKl/ERKZ, JNK2/JNK1, p38; 1:1,000 for phospho-AKT-ser473 and phospho-AKT-thr308) in blocking buffer (5 % BSA in TBST) with gentle shaking overnight (~18 h) at 4°C. The primary antibodies were removed and the membranes were washed 4 times with TBST and incubated with horseradish peroxidase-conjugated anti-rabbit IgG secondary antibody (Cell signaling, Beverly, MA), (1 :20,000 for phospho-ERKI/ERK2, phospho-JNKZ/JNKI, phospho-p38 and ERKl/ERKZ, JNK2/JNK1, p38; 1:2,000 for phospho-AKT-ser473, phospho-AKT- thr308) for 1 h at room temperature and detected with western blotting chemiluminescence luminol reagent (Santa Cruz Biotechnology Inc., Santa Cruz, CA) 109 and/or Supersignal West Dura Extended Duration Chemiluminescent Substrate (Pierce Biotechnology Inc., Rockford, IL). Membranes were exposed to X-ray film for 1-10 min. Densitometry. Density of bands was estimated using the GelExpert (San Mateo, CA) or Kodak ID 3.6 computer software program (Kodak Scientific Imaging System, New Haven, CT) and are expressed in % of time zero. Then each value for Phosphorylated forms of ERKl, ERK2, JNK2, JNKl, and p38 was divided by the corresponding control for total ERKl, ERK2, JNK2, JNKl , and p3 8, respectively. 110 RESULTS To determine effects of sphinganine on the status of protein expression and/or phosphorylation/activation of ERKl/ERKZ, JNKZ/INKl, p38, AKT-ser473, AKT- thr308, sub-confluent HT-29 cells were cultured with sphinganine at 35 pM, which promotes apoptosis (Ahn and Schroeder, 2002b). Cellular protein was isolated and kinases were evaluated using Western blot analysis. The density of bands was calculated using densitometry and expressed in % of time zero and then each value for phosphorylated forms of ERK], ERK2, JNK2, JNK], and p38 was divided by the corresponding control for total ERK], ERK2, JNK2, JNK], and p3 8, respectively. Sphinganine has small efl'ects on early activation of ERK] and ERKZ. Sphinganine at 35 pM has small effects on the increase of active phosphorylated forms of ERKl/ERK2 (p44/p42) at 15, 30, and 60 min compared to the corresponding control, and had little or no effect on protein expression levels of ERKl/ERKZ (Figure 4.1). Sphinganine causes early activation of .flVKZ and JNK]. Phosphorylated forms of JNK2/JNK1 were virtually undetectable in controls (Figure 4.2). Sphinganine at 35 pM clearly increased the active phosphorylated forms of JNK2/JNK1 (p54/p46) in time- dependent manner compared to the corresponding control, while having little or no effect on protein expression levels of JNK2/JNK1. The strongest activation/phosphorylation of JNK2/JNK1 was observed at 60 min (Figure 4.2). Sphinganine causes early activation of p38 mitogen activated protein kinase. Phosphorylated forms of p38 mitogen activated protein kinase were virtually undetectable in controls (Figure 4.3). Sphinganine at 35 pM clearly increased the active 111 phosphorylated forms of p38 at 15, 30, and 60 min compared to the corresponding control, but had little or no effect on protein expression levels of p38. The strongest activation/phosphorylation of p38 was observed at 60 min Giigure 4.3). Sphinganine causes early inhibition of AK T-ser4 73 and AK T -thr308. Sphinganine at 35 pM (an apoptotic concentration) inhibited the active phosphorylated forms of AKT-ser473 at 60 min and AKT-thr308 at 15 and 60 min compared to the corresponding control (Figure 4.4). 112 Minutes 0 15 15 30 30 60 60 Sphinganine - — + — + — + - ._.. _— .._. ~.. P-ERK1(p44) 8.. - - "" "' P-ERK2(p42) Minutes 0 15 15 30 30 60 60 Sphinganine — .— + — + — + ""‘ W “H - ERK1(p44) .. ERK2(p42) a: e a: g E 3 1-8 ‘3 8 t8 SA 5 o 3 0 :2 g 12 g g 1.2 ‘- ... u- :1“ a: o" ' control g 3: 0'6 control 0'. v o 1 . i v o o 15 30 45 so Minutes 3 1‘5 3b is so Minutes Figure 4.1. Effects of sphinganine on activation of extracellular signal- regulated kinase 1/2 (ERK1/ERK2) in HT -29 human colon cancer cells. Sub-confluent cells were treated without (O) or with sphinganine (0, SA) at 35 pM for 0, 15, 30, and 60 min. Cellular proteins were isolated, quantitated via Bradford assay, and analyzed via Western blotting. The density of bands was calculated using Kodak ID 3.6 computer software program and expressed in % of time zero and then each value for P-ERK1 and P-ERK2 was divided by the corresponding control for total ERK1 and ERK2. 113 Time(min) 0 15 15 30 30 60 60 Sphinganine - - + - + — + f - -- -- P-JNKZ (p54) L ' P-JNK1 (p46) Time (min) 0 15 15 30 30 6O 6O Sphinganine - -__-_ + - * + .. + ..... = ~ 9 #- .. "r M JNK2 (p54) . ~ JNK1 (p46) a -m- s J“- z 0 Z 2 '1 '5 150 - 2 9’ 150 * SA 3 N a 3 o 0 g 100 E 5 “Mr E “5 5o .. g '3 50 .. § >5 7 23 control a o o. o 0 15 30 45 60 0 15 30 45 60 Minutes Minutes Figure 4.2. Sphinganine causes early activation of c-Jun NHz-tenninal kinase-2H (JNK2/JNK1) in HT-29 human colon cancer cells. Sub- confluent cells were treated without (O) or with sphinganine (0, SA) at 35 pM for 0, 15, 30, and 60 min. Cellular proteins were isolated, quantitated via Bradford assay, and analyzed via Western blotting. The density of bands was calculated using Gel Expert and Kodak D 3.6 computer software program, respectively and expressed in % of time zero. Then each value for P-JNKZ and P-JNK1 was divided by the corresponding control for total JNK2 and JNK1. 114 Time (min) 0 15 15 30 30 60 60 Sphinganine. _ _ + _ + .. -r- ' _m" P-p38 0 15 15 30 30 60 60 Time (min) — — + — f — + Sphinganine _ 1 -..... ¢ . - Q . p38 P-p3BITotal p38 (“lo of time zero) a s E E E ‘2‘ 0 15 30 45 60 Minutes Figure 4.3. Sphinganine causes early activation of p38 mitogen activated protein kinase in HT-29 human colon cancer cells. Sub- confluent cells were treated without (0) or with sphinganine (0, SA) at 35 pM for O, 15, 30, and 60 min. Cellular proteins were isolated, quantitated via Bradford assay, and analyzed via using Western blotting. The density of bands for P-p38 and total p38 was calculated using Gel Expert and Kodak ID 3.6 computer software program, respectively and expressed in % of time zero. Then each value for P-p38 was divided by the corresponding control for total p38. 115 Time (min) Sphinganine - 1...... .§--_-— P-AKT-cer473 Time (min) Sphinganine - t . P-AKT-ser473 (% of time zero) a s s ii § 5 Figure 4.4. Sphinganine causes early inhibition of AKT-ser473 and AKT- thr308 in HT-29 human colon cancer cells. Sub-confluent cells were treated without (0) or with sphinganine (0, SA) at 35 pM for 0, 15, 30, and 60min. Cellular proteins were isolated, quantitated via Bradford assay, and analyzed using Western blotting. The density of bands was calculated using Kodak ID 3.6 computer software program and expressed in % of time zero. 15 + 15 0 15 15 «1- 0 i ii l P )- contro SA 1 l J c 15 so 45 eh Minutes 116 30 30 30 + 30 P-AKT-thr308 (% of time zero) 1- 60 60 60 -— 'l- are? P-AKT-thr308 150 12° ' control 90 r so .- 3“ ’ SA 0 l L l L L o 15 30 45 60 Minutes 60 4. DISCUSSION Sphingoid bases (sphingosine and sphinganine) and Cz-cerarnide inhibit growth and induce apoptosis in HT-29 and HCT—l 16 human colon cancer cells, with sphinganine being the most potent (Ahn and Schroeder, 2002b). One possible mechanism by which sphinganine inhibits proliferation and induces apoptosis is by altering the activation of MAPKs and AKT. The current study is the first to demonstrate that sphinganine at an apoptosis-inducing concentration (35 pM) causes early inhibition of AKT and early and strong activation of JNK, and p38 and in HT-29 human colon cancer cells. Activation of JNK and p38 is observed as early as 15 min and is maintained up to 60 min. Though ERK activation has most often thought of as mitogenic and anti- apoptotic, recent evidences suggest that ERK activation is not always required for mitogenic (Thamileselvan et al. 2002) and anti-apoptotic (Chang et al. 2001b) effects. In the present study, sphinganine had a small effect on ERK activation. The relatively greater activation of ERKl/ERKZ at 15 min for both control cultures and sphinganine- treated cultures compared to other groups at 0, 30, and 60 min might be associated with the “burst” of sphinganine and sphingosine that occurs when cells in culture are changed to flesh medium (Smith et al, 1995). Our results contrast with those previously reported by Kim et a1 (1999) who found that 50 pM Cz-ceramide, an acylated sphingoid base, inhibited ERK activity in HT-29 cells as early as 15 min and inhibition was sustained for up to 3 h. Differing culture conditions (e.g. 1% F BS in the present study versus 10 % FBS in the earlier study by Kim et al. 1999) may have contributed to the disparity in results between the studies. 117 We found that sphinganine causes strong activation of JNK at 15, 30, and 60 min in HT-29 human colon cancer cells. Our results are consistent with a previous study that showed that Cz-ceramide increases JNK activity in HT-29 cells (Kim et al. 1999). Our study demonstrated that sphinganine causes maximum activation of JNK at ~60 min; whereas, the previous study showed Cz-ceramide causes maximum activation of JNK at ~15 min (Kim et al. 1999). Jarvis et al (1997) showed that sphingosine-mediated lethality of U937 human monoblastic leukemia cells is largely associated with weak stimulation of JNK2/JNK1 and enhanced p38 mitogen activated protein kinase activity (Jarvis et a1. 1997). It is noteworthy that several chemotherapeutic drugs including indomethacin (a nonsteroidal anti-inflammatory drug) (Kim et al. 1999) and tamoxifen (Mandlekar et al. 2000) cause turnover of complex sphingolipids to bioactive sphingolipid metabolites, such as ceramide and/or sphingoid bases, and induce apoptosis by stimulating JNK activity. Our findings might suggest that the sphingoid base sphinganine might mediate the apoptotic effect of chemotherapeutic drugs in cancer cells, in part, by activating JNK. Alternatively, sphingosine could induce apoptosis by inhibiting AKT (protein kinase B). AKT is a cytoplasmic serine/threonine kinase that promotes cell survival and inhibits apoptosis (for review: Testa and Bellacosa, 2001, Talapatra and Thompson, 2001). Enhanced AKT activity is associated with cancer progression in various tissues (Graff et al. 2000, Krasilnikov, 2000, Tsatasanis and Spandidos, 2000, Yuan et al. 2000, Sun et al. 2001). AKT activation is achieved by phosphorylations at ser473 and thr308 residues and thr-308 phosphorylation is necessary for AKT activation and ser473 phosphorylation is only required for maximal activity (Testa and Bellacosa, 2001). 118 However most studies have examined phosphorylation status of AKT only at ser473. The present study demonstrated that sphinganine causes early inhibition of the phosphorylated/active forms of AKT-ser473 and AKT-thr308 in HT-29 human colon cancer cells. Our findings are consistent with those of Chang et al (2001b) who showed that sphingosine induced apoptosis and suppressed activation of AKT. Moreover, apoptosis induced by sphingosine was attenuated in cells transfected with constitutively active AKT (Chang et al. 2001b). Thus, sphinganine and/or sphingosine might induce apoptosis, in part, by inhibiting AKT activation in human cancer cells. In conclusion, this is the first study to demonstrate that sphinganine at a concentration-inducing apoptosis causes early and strong activation of JNK and p3 8, with minimal effect on ERK activation, and early inhibition of AKT in HT-29 human colon cancer cells. The findings suggest that JNK and p38 mitogen activated protein kinase and AKT might mediate the action of sphinganine to initiate apoptosis of colon cancer cells. 119 Acknowledgement We express our thanks to: Dr. Yon-co Chung for technical advice on western blot analysis; Mr. Chris Salvadore at Cell Signaling Co. for providing a sample of phopho- JNK antibody; Drs. Dale R. Romsos and Chia—Cheng Chang for their encouragement; Dr. James Pestka for providing access to his experimental instruments. 120 CHAPTER 5 Evaluation of sphinganine and sphingosine as breast cancer chemotherapeutic and chemopreventive agents using human breast stem, normal, and tumorigenic cell models Running title: Sphingoid bases and breast cancer Key words: sphinganine; sphingosine, cancer, stem cells, apoptosis, differentiation 1This chapter is prepared for publication with authors Eun Hyun Ahn, Chia-Cheng Chang, and Joseph J. Schroeder. 121 ABSTRACT Sphingolipids have gained attention for their potential as chemotherapeutic or chemopreventive agents based on in vitro cancer cell and animal studies. However, no comparative study of the effects of the sphingolipid metabolites sphingosine and sphinganine on proliferation and differentiation in normal human epithelial cells versus tumorigenic cells and epithelial stem cells has been reported. The purpose of this study was to evaluate the chemotherapeutic and chemopreventive potential of sphingoid bases using a novel cell culture system of normal human breast epithelial cells (HBEC) developed from tissues of healthy women obtained during reduction mammoplasty (type I HBEC with stem cell characteristics and type II HBEC with basal epithelial cell phenotypes) and transformed tumorigenic type I HBEC. The results show that sphinganine more potently inhibited the growth and induced apoptosis of tumorigenic type I HBEC than sphingosine (ICso for sphinganine 4 pM; sphingosine 6.4 pM). High concentrations of either sphingoid base (8-10 pM) arrested the cell cycle at G2/M with reductions in S and Go/Gl phases, while sphingosine showed a threshold effect. Sphinganine also more potently inhibited the growth and caused death of type I HBEC than sphingosine, thus sphingoid bases may reduce target cells for breast carcinogenesis. In comparison, type II HBEC were less sensitive to the growth inhibitory effects of sphingoid bases than tumorigenic type I HBEC and type I HBEC, suggesting that sphingoid bases may serve as chemotherapeutic agents. At concentrations (0.05-0.5 pM), which are below the growth inhibitory ranges, sphingoid bases induced differentiation of type I HBEC to type H HBEC, as detected morphologically and via expression of a novel tumor suppressor protein maspin in type II HBEC. The current study is the first to 122 systematically compare the chemotherapeutic and chemopreventive properties of sphingoid bases in normal, stem, and tumorigenic breast cells. The data support the potential for sphingoid bases to be employed as chemotherapeutic and chemopreventive agents against human breast cancer. 123 INTRODUCTION Breast cancer is the most common cancer among women in the United States and worldwide (American Cancer Society 1999). Although the etiology of breast cancer is not well understood, the cumulative exposure of breast tissue to estrogen appears to be a major factor in the development of breast cancer risk (Henderson et al. 1993, Jordan and Morrow 1999). Recently the identification of protective dietary compounds that prevent the development of diseases including breast cancer has gained much attention. In particular, the search for and development of chemotherapeutic and/or chemopreventive agents that do not exert side effects and cause drug resistance has been an active area of research. Sphingolipids are constituents of dairy and soy products at relatively high concentrations (Ahn and Schroeder, 2002a). These dietary sphingolipids are digested and absorbed as ceramide and sphingoid bases (sphingosine and sphinganine) (Schmelz et al. 1994). Ceramide and sphingoid bases function as second messengers in signal transduction pathways, inhibiting cell proliferation and inducing apoptosis (programmed cell death) in a variety of human cancer cells (Ahn and Schroeder 2002b, Sweeney et al. 1996) including breast cancer cells (Gill et al. 1997, Cai et al. 1997). For example, in HT-29 and HGT-116 human colon cancer cells sphinganine more potently induces apoptosis than sphingosine (Ahn and Schroeder 2002b). One of the mechanisms by which many clinically important drugs, including antiestrogens such as tamoxifen and the novel synthetic retinoid N—(4-hydroxyphenyl) retinamide (4-HPR) that inhibit proliferation and induce apoptosis of cancer cells, is through activation of sphingolipid- mediated pathways (for review: Senchenkov et al. 2001). 124 Most studies examining sphingolipids have focused on the pro-apoptotic actions of ceramide (acylated form of sphingoid bases) and fewer studies have examined the actions of sphingosine and sphinganine (Figure 5.1). Furthermore, it is uncertain whether the sphingosine or sphinganine backbones of ceramides mediate anti-proliferative and pro-apoptotic activities of ceramide on cancer cells (Sakakura et al. 1998, Back et al. 2001, for review: Goswami and Dawson, 2000, Radin, 2001). In addition, no study has compared the effects of sphingoid bases on proliferation and differentiation in human normal epithelial cells to that in tumorigenic cells and epithelial stem cells. Previously, we have reported the characterization of two types of morphologically distinguishable and phenotypically different normal human breast epithelial cells (HBEC) from reduction mammoplasty tissues of healthy females (type I and type II HBEC) (Kao et al. 1995, for review: Chang et al. 2001a) (Figure 5.2). Type I HBEC display stem cell characteristics based on observations that type I HBEC differentiate to type II HBEC and mixture of type I and type II HBEC form budding and ductal organoids on Matrigel (Kao et al. 1995, Chang et al. 2001a). Type I HBEC express estrogen receptors (Kang et al. 1997) and are more susceptible to neoplastic transformation, telomerase activation, and immortalization (Kao et al. 1995, Kao et al. 1997, Kang et al. 1998, Sun et a1. 1999). Therefore type I HBEC appear to be the major target cells for breast carcinogenesis. Type H HBEC do not express estrogen receptors, rarely become immortal after simian virus 40 transformation and, unlike type I HBEC, do not have the capability of anchorage independent growth (Kao et al. 1995, Sun et al. 1999, Chang et al. 2001a). In the current study, this novel culture system of type I and type H HBEC was used as a cell model system to assess the chemopreventive potential of sphinganine in 125 OH "'13" Sphingosine H cuon NH3+ Sphinganine OH /W\/\/\/\/\/J\/CH20H \ \/\/\/\/\/\/\/\/\/NH Ceramide 3 OH MCI-'20“ H3C\/NH C2-Ceramide 41 OH H3C\,NH Cz-dihydroceramide ll Figure 5.1. Structures of sphingosine, sphinganine, ceramide, Cz-ceramide, and Cz-dihydroceramide. 126 Type I HBEC (stem cells) Type II HBEC Differentiation 7 f— SV40 transfection ——i ”133‘", lmmortalized at Immortallzed at . low frequency 3 high frequency X-ray 7 M13SV1 R2 Weakly tumorigenic (— C-erbBZ/neu oncogene N V M13SV1R2N1 Highly tumorigenic Tumorigenic type I HBEC Figure 5.2. Derivation of tumorigenic type I HBEC and type ll normal human breast epithelial cells (HBEC) from type i HBEC with stem cell characteristics. Abbreviation used: SV40, simian virus 40. 127 comparison to sphingosine. The chemotherapeutic potential of sphinganine and sphingosine was also evaluated by examining the effects of the sphingoid bases on tumorigenic type I HBEC representing breast tumor cells in comparison to type II HBEC representing normal breast cells. The following criteria were applied to determine the chemotherapeutic and chemopreventive properties of sphingoid bases in tumorigenic type I HBEC, and type I and type II HBEC: 1) A chemotherapeutic agent is expected to inhibit the proliferation and induce apoptosis of tumorigenic type I HBEC; 2) A chemopreventive agent is expected to preferentially inhibit the proliferation and induce the differentiation of type I HBEC; thereby, decreasing the target stem cells for neoplastic transformation. The objectives of this study were to determine: 1) whether sphingoid bases preferentially inhibit proliferation of tumorigenic type I HBEC and type I HBEC, compared to type H HBEC; 2) the effects of sphingoid bases on apoptosis and the cell cycle in tumorigenic type I HBEC; and 3) whether sphingoid bases induce the differentiation of type I HBEC to type II HBEC. 128 MATERIALS AND METHODS Sphingolipids. Sphinganine (D-erythro-dihydro-sphingosine) was obtained from Matreya Co. (cat. no. 1831, State College, PA) and D-erythro-sphingosine was from Sigma-Aldrich Co. (S-6879, St. Louis, MO). Stock solutions of sphinganine and sphingosine were prepared by dissolving the sphingoid base in ethanol at 50 mM and storing at 4°C. Working solutions of sphingoid bases were prepared as 1:1 complexes with 1 mM bovine serum albumin (BSA) in phosphate buffered saline (PBS) and stored at 4°C up to one month. Prior to experiments, sphingoid bases were incubated at 37°C overnight and sonicated for 2 h to ensure sphingoid bases-BSA complex formation. HBEC culture media. The MSU-1 medium (Kao et al. 1995) with various supplements was used to culture type I HBEC, tumorigenic type I HBEC, and type H HBEC. The MSU-1 medium is a 1:1 mixture of a modified Eagle’s MEM (called D medium) (Invitrogen Co., Carlsbad, CA) (Chang et al. 1981) and a modified MCDB 153 (M-7403, Sigma-Aldrich Co., St. Louis, MO) (Pittelkow et al. 1986), supplemented with human recombinant epidermal growth factor (0.5 ng/mL, E-9644, Sigma-Aldrich Co.), insulin (5 pg/mL, I-1882, Sigma-Aldrich Co.), hydrocortisone (5 pg/mL, H-0888, Sigrna- Aldrich Co.), human transferrin (5 pg/mL, T-7786, Sigma-Aldrich Co.), l7-B-estradiol (10'8M, 13-2257, Sigma-Aldrich Co.), Gentamicin (50 pg/mL, 15710-064, Invitrogen Co.) (Kao et al. 1995). Development of human breast epithelial cells (HBE C) from reduction mammoplasty tissues of healthy females. Breast tissues of healthy women at 23, 26, and 23 years of age were obtained during reduction mammoplasty at a hospital in Lansing, MI. These initial primary HBEC cultures developed from breast tissues were 129 designated as HME23, HME29, and I-IME30, respectively. Kao et a1 (1995) and Chang et al (2001a) developed HBEC from these specimens. Procedure for the development of type I HBEC with stem cell characteristics and type II HBEC from reduction mammoplasty tissues has been described (Kao et al. 1995, Trosko et al. 2000). HBEC were stored in liquid nitrogen until use. Separation of type I HBE C with stem cell characteristics and type II HBEC. HBEC stored at liquid nitrogen were thawed at 37°C and plated in the MSU-1 medium supplemented with 5 % PBS in a 100 mm dish, designated as “Plate A”. After 2 h incubation, the unattached cells were transferred to a 15 mL tube and the MSU-1 medium supplemented with 0.4 % bovine pituitary extracts was added to the attached cells in Plate A. After centrifugation (1000 rpm for 8 min) of the unattached cells, the supernatant was removed and the cell pellet was suspended in the MSU-1 medium supplemented with 0.4 % bovine pituitary extracts and plated in a dish labeled “Plate B” with the MSU-1 medium supplemented with 0.4 % bovine pituitary extracts. Both plates of cells were cultured overnight at 37 °C in incubators supplied with 5% C02 and humidified air. The next day, the unattached cells in the medium in plate B were collected in a 15 mL tube and MSU-l medium supplemented with 0.4 % bovine pituitary extracts was added to the attached cells in plate B. After centrifugation (1000 rpm for 8 min) of the medium containing the unattached cells from plate B, the supernatant was removed and the cell pellet was suspended in the MSU-l medium supplemented with 5 % F BS and plated in the MSU-1 medium supplemented with 5 % F BS in a dish labeled “Plate C”. Plate A contained fibroblasts and was not used for experiments. Plate B contained 130 mainly type II HBEC. Plate C contained mainly type I HBEC and a few fibroblasts (which, after forming colonies, were marked and removed by scraping with a rubber policeman. Appendix L: Figure A.13-photographs). Derivation of in vitro neoplastically transformed HBEC lines from HBEC, normal human mammary epithelial culture from reduction mamoplasty tissues. Previously Kao et a1 (1995) and Kang et a1 (1998) derived in vitro neoplastically transformed HBEC lines (Ml3SVl, M13SV1R2, and M13SV1R2N1) from type I HBEC (for review: Chang et al. 2001a). These transformed type I HBEC were stored in liquid nitrogen until use. Type I HBEC with stem cell characteristics developed from reduction mammoplasty tissues were sequentially transformed to immortalized/non-tumorigenic cells (MlBSVl), weakly tumorigenic cells (Ml3SV1R2), and highly tumorigenic cells (M13SV1R2N1) by transfections with SV-40, X-ray, and neu oncogene (Kao et al. 1995, Kang et al. 1998). In the current study, transformed tumorigenic type I HBEC (M13SV1R2N1) were used as the breast cancer cell model (Figure 5.2). Culture of type I HBEC, tumorigenic type I HBEC, and type II HBEC. Type I HBEC and tumorigenic type I HBEC were cultured in the MSU-1 medium supplemented with 5 % FBS (Kao et al., 1995). Type H HBEC were cultured in the MSU-1 medium supplemented with 0.4 % bovine pituitary extracts (prepared from bovine pituitary glands, Pel-Freez Biologicals, Rogers, AR) (Kao et al. 1995). Cells were cultured at 37 °C in incubators supplied with 5% C02 and humidified air. Assessment of cell proliferation. To assess the effects of sphingoid bases 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. Briefly, confluent cells were trypsinized and 131 cells were seeded at a density of 6 x 104 cells per well in 6-well dishes (i.e. 6.3 x 103 cells/cm2 growth area) and cultured in 2 mL of the MSU-1 medium supplemented with 5 % FBS for 24 h for tumorigenic type I HBEC to insure that cells were in log phase before treatment with sphingoid bases. For type I HBEC, cells were cultured in the MSU-1 medium supplemented with 5 % F BS for 3 d since type I HBEC have lower cell plating efficiency and proliferation rate. For type II HBEC, cells were cultured in the MSU-1 medium supplemented with 5 % F BS for 1 d and then switched to the MSU-1 medium supplemented with 0.4 % bovine pituitary extracts for 2 d since type II HBEC are sensitive to trypsinization and have lower cell plating efficiency after re-plating in the MSU-1 medium supplemented with 0.4 % bovine pituitary extracts. Then these media were replaced with FBS-free MSU-l medium. Various concentrations of sphinganine and sphingosine were added directly to each dish and the cells were cultured for up to 6 days. Fresh FBS-free MSU-1 medium and treatments were renewed after 3 (1. Following treatments, the media and floating dead cells were removed, the viable attached cells were rinsed with 1 mL of PBS, and the cells were lysed with 1 mL of 0.1 M NaOH. Total nucleic acids concentrations were determined by measuring the absorbance of the cell lysate at 260 nm using Beckman DU-7400 spectrophotometer (Beckman Coulter Inc., Fullerton, CA). Flow cytometry of cell cycle and population. Flow cytometric analysis was performed as described by Telford et al. (1994) with modifications. Confluent tumorigenic type I HBEC were trypsinized, seeded at a density of 3.5 x 105 cells/ 100 mm dish (i.e. 6.3 x 103 cells/cm2 growth area), and cultured with 5 mL of the MSU-1 medium supplemented with 5 % FBS for 24 h to ensure that cells were in log phase before 132 treatment with sphingoid bases. Then the medium was replaced with FBS-free MSU-l medium. Various concentrations of sphinganine and sphingosine were added directly to each dish and the cells were cultured for 12 or 24 h. After treatments, floating cells were collected and 1.5 mL PBS with 8% F BS (heat inactivated at 50°C for 30 min and filtered with 0.22 pm filter syringe) was added to each tube. Attached cells were trypsinized for 10 min in the incubator at 37°C and supplied with 5 % C02 and humidified air. Trypsinization was terminated by adding 3 mL of PBS containing 4 % FBS to each dish and detached cells were transferred to the previous tubes containing floating cells. After centrifugation (1200 rpm, 4 min), the supernatant was removed and PBS (pH 7.4) containing 50 % FBS was added. Cells were fixed with 70 % cold ethanol at a final concentration of 50-53 %. After storage at 4°C for 6-8 h, cells were pelleted by centrifugation (1200 rpm, 4 min) and resuspended in 500 pL of PBS containing 4 % FBS. Then, 300 pL DNA staining solution [4 % FBS, 0.1 % Triton X-100, 100 pM EDTA (pH 8.0), 0.05 mg/mL RNase A (50 units/mg), 50 pg/mL propidium iodide in PBS (pH 7.4) containing 4% FBS] was added and the cells were kept at 4°C for 2 h and then filtered through 40 pm nylon mash (Small Parts Inc., Miami Lakes, FL) and collected into Falcon tubes. The cells were kept at 4°C for 3-5 h before reading using a FACS Vantage Flow Cytometer (Becton Dickinson, San Jose, CA). The total number of cells analyzed for each sample was 7000. The percentages of cells in Go/Gl, S, and G2/M phases were determined using Mod-fit LT version 3.1 software program (Verity Software House, Topsham, ME). The Mod-fit software program does not include any cell populations outside of standard cell cycle populations when calculating areas, thus excluding sub-Go/Gl cells. The F CS 133 Express version 2.0 software (De Novo Software, Thomhill, Ontario, Canada) was used to determine the percentage of apoptotic cells represented by the A0 peak in the hypo- diploid (DNA content less than 2N) sub-Go/01 area to the left of the Go/Gl diploid (DNA content 2N) peak. Assessment of type I HBE C difierentiation. The effects of sphingoid bases on the ability of type I HBEC to differentiate to type H HBEC were evaluated by counting morphologically distinguishable colonies of type I, type H, and type I surrounded by type H HBEC. Type I HBEC at cell seeding density of 5000 cells/60 mm dish with grids (i.e. 238.1 cells/cm2 growth area, 1250 cells/mL) were grown in 4 mL of the MSU-1 medium supplemented with 5 % FBS (which supports the growth of type I HBEC, but not type H HBEC) for 2 d. On the third day, cells were cultured in FBS-free MSU-1 medium (which supports growth of both type I and type H HBEC) and then sphingoid bases at various concentrations and cholera toxin (C-8052, Sigma-Aldrich Co.) as a positive control (Kao et al., 1995) were directly added to each dish. Cells were cultured in FBS-free MSU-1 medium for 7 days with changing of fresh FBS-free MSU-1 medium and renewed treatments on day 3 and day 5. At the end of the experiment, the medium containing floating cells was removed. Cells were washed once with PBS and stained with 1% crystal violet (Sigma-Aldrich Co.) for 2 min and then rinsed with water. Stained colonies in dishes were air-dried and kept at room temperature. The number of colonies of type I, type H, and type I surrounded by type H HBEC (the cell culture started from single cell plating of pure type I HBEC) were visually identified and counted via microscopy. To ensure objectivity, the identities of the treatment for each dish were unknown (blinded) to two people who were instructed to 134 score colonies and did not participate in other parts of the differentiation experiment. The same dishes were independently counted by two people with the same criteria and the results were consistent to each other. Immunofluorescence staining. Type I HBEC at cell seeding density of 1250 cells per well in 4-well chamber slide (i.e. 735.3 cells/cm2 growth area) were grown in 1 mL of the MSU-1 medium supplemented with 5 % FBS (which supports the growth of type I HBEC, but not type H HBEC) for 2 d. On the third day, cells were cultured in FBS-free MSU-l medium (which supports growth of both type I and type H HBEC) and then sphingoid bases at various concentrations were directly added to each dish. Cells were cultured in FBS-free MSU-1 medium for 7 days with changing of flesh FBS-free MSU-1 medium and renewed treatments on day 3 and day 5. At the end of the experiment, the medium containing floating cells was removed. After fixation of cells with 4 % paraformaldehyde in PBS for 30 min at room temperature with gentle shaking, cells were rinsed with PBS and permeabilized with 0.5 % triton X-100, 2% BSA, 0.05 % NaN3 in PBS. Permeabilization reagent is removed and cells were incubated with a mouse anti-human maspin monoclonal antibody (1:100 dilution, cat no. 554292, BD Biosciences-Pharmingen, San Diego, CA) for 20 h at 4°C with gentle shaking. After rinsing cells with PBS, cells were incubated with FITC-conjugated sheep anti-mouse IgG (1:200 dilution, cat no. 115-095-166, Jackson ImmunoResearch Laboratories Inc., West Grove, PA) for 45 min at 4°C. After rinsing cells four times with PBS, cells were mounted with Aqua-Poly mount (Polysciences Inc., Wanington, PA) and photographed using a fluorescence/phase contrast microscope equipped with a digital camera (Diagnostic Instruments, Sterling Heights, MI). 135 Statistical analyses. Data for total nucleic acids assay at various concentrations and culture periods were analyzed by two-way factorial analysis of variance (AN OVA). After application of two-way AN OVA, the significance of differences in the means between control and treatment groups at specific culture periods was evaluated by multiple comparisons using the Bonferroni method. Data for total nucleic acids at various concentrations and a single culture period were analyzed by one-way factorial AN OVA. After application of one-way AN OVA, the significance of differences in the means between control and treatment groups at a single culture period was evaluated by multiple comparisons using the Student-Newman-Keuls method. Differences were considered significant at p < 0.05. 136 RESULTS Sphinganine more potently inhibits the growth and causes death of tumorigenic type I HBE C than sphingosine. To determine the effects of sphinganine on growth and death of tumorigenic type I human breast epithelial cells (HBEC) in comparison to sphingosine, sub-confluent cells were cultured with sphinganine and sphingosine at various concentrations and total nucleic acids were determined as an index of cell number (Figme 5.3; Appendix K: Figure AID—photographs). Total nucleic acids in control cultures doubled within 1 day and increased 10 fold in 5 days (Figure 5.3-Lefi panel). Sphinganine caused concentration- and time-dependent decreases in proliferation of tumorigenic type I HBEC. Specifically, addition of sphinganine at 3.5 and 5 pM significantly reduced total nucleic acids at l d by 53 and 89 %, at 3 d by 49 and 89 %, and at 5 d by 66 and 95 %, respectively compared to the corresponding controls (p<0.05). In contrast, sphingosine at 5 pM significantly reduced total nucleic acids at 1 d by 51 %, at 3 d by 35 %, and at 5 d by 36 % compared to the corresponding controls (Figure 5.3- Lefi panel). These data suggest that sphinganine more potently inhibited the growth and caused death of tumorigenic type I HBEC than sphingosine. The effects of sphingoid bases on death of tumorigenic type I HBEC were further examined at concentrations ranging from 2 to 10 pM for 24 b (Figure 5.3-Right panel; Figure 5.4-Photographs). Sphinganine at 2, 3.5, and 5 pM decreased total nucleic acids by 24, 37, and 85 %, respectively compared to the control. Sphinganine at 8 and 10 pM completely killed cells. In contrast, sphingosine at 3.5, 5, and 8 pM decreased total nucleic acids concentrations by 7, 23, and 78 %, respectively compared to the control. 137 Tumorigenic type I HBEC A 180 2 100° Sphinganine 150 o 800 g N 120 ., 600 1- .§ 400 3 A 9° : zoo a 2 6° 2 0 fl 3 3° 0 0 i; o 1 3 5 “‘5’ ° _ 8_ 1: Days .2 '5 pM Sphinganine E 1000's hin osine ‘7', 3 18° .2 300- p g E :35150 2 , - 120 3 60° * ‘3 so : 400) l- 60 E 200» so 0 o l I I 4 1 1 '— o 1 3 5 ”o 2 4 e 3 1o Days pM Sphingosine Figure 5.3. Sphinganine more potently inhibits the growth and causes the death of tumorigenic type I HBEC than sphingosine. Lefl panel: Sub- confluent cells were cultured with sphinganine at 0 (e), 0.5 (O), 2 (V), 3.5 (V), and 5 M (D) and with sphingosine at O (c) and 5 pM (D) for 1, 3, and 5 days. Right panel: Sub-confluent cells were cultured with sphinganine and sphingosine at O, 2, 3.5, 5, 8, and 10 pM for 24 hours. Total nucleic acids were measured as an index of cell number. Results are expressed as a percentage of the zero hour. Data shown are mean 1 SEM (n=3). Where an error bar is not shown, it lies within the dimensions of the symbol. Means at each culture period with an asterisk (*) are significantly different (P < 0.05) from the corresponding control. 138 Sphinganine Tumorigenic type I HBEC at 24 h Control .. . , 10pM Figure 5.4. Sphinganine more potently inhibits the growth and causes death of tumorigenic type | HBEC than sphingosine at 24 h (photographs). Sub-confluent cells were cultured with sphinganine and sphingosine at 0, 2, 3.5, 5, 8, and 10 pM for 24 hours. Magnification 10X plus 10X. Images in this dissertation are presented in color. 139 Sphingosine at 10 pM completely killed cells (Figure 5.3-Right panel). The ICso for sphinganine was approximately 4 pM, while that for sphingosine was ~ 6.4 pM. Sphinganine more potently induces apoptosis of tumorigenic type I HBE C than sphingosine. Both sphingoid bases arrest the cell cycle at G/M with reductions in S and 61/62 phases. The effects of sphingoid bases on cell cycle and apoptosis of tumorigenic type I HBEC were analyzed via flow cytometry using concentrations of sphingosine and sphinganine found to be growth inhibitory and cytotoxic in total nucleic acids assays (Figure 5.3). As shown in Figure 5.5, sphinganine at 8 pM increased the number of cells in the hypo-diploid sub-Go/Gl region, indicative of apoptosis (called A0). To quantify the degree of apoptosis, cells in the A0 peak were quantitated and expressed as a percentage of the total cell population. Specifically, about 4.3 % of the control cells for sphinganine were in the A0 apoptotic peak, while sphinganine at 3.5, 5, and 8 pM increased the number of apoptotic cells to 7.9, 8.6, and 10.9 %, respectively (Figure 5.6). About 2.5 % of the control cells for sphingosine were in the A0 apoptotic peak. In comparison, sphingosine at 5, 8, 10 pM increased the number of apoptotic cells to 3.4, 4.3, and 7.8 %, respectively (Figure 5.6). The effects of sphingoid bases on the cell cycle distribution of tumorigenic type I HBEC were also examined (Figure 5.6). The percentages of cells in Go/Gl, S, G2/M after 24 h exposure to each treatment were: control for sphinganine (34; 39; 27), 3.5 pM sphinganine (41; 40; 20), 5 pM sphinganine (39; 37; 24), 8 pM sphinganine (29; 24; 47), control for sphingosine (36; 38; 26), 5 pM sphingosine (36; 36; 28), 8 pM sphinganine (29; 35; 37), 10 pM sphingosine (27; 36; 37). Sphinganine at 8 pM and sphingosine at 8 and 10 W for 24 h increased the percentages of cells in G2/M and decreased the 140 percentages of cells at S and Go/Gl phases compared to the corresponding controls (Figure 5.6). Sphingosine at 8 pM for 12 h also caused accumulation of cells at 02M and reductions at S and Go/Gl phases (Appendix K-Figure A.11). Interestingly sphingosine at both 8 and 10 pM caused similar degrees of accumulations of cells in G2/M, although 10 pM sphingosine caused a much greater increase of apoptotic cells than 8 pM sphingosine (Figure 5.6). This implies that sphingosine has a threshold greater than 5 but less than 8 pM which causes accumulation of cells in G2/M phase (Figure 5.6). 141 120 Tumorigenic type I HBEC 100 1 Control 120 100 Sphinganine sol car/e, 60 I A9 I W 20 l ' 11W ““111wa 0 160 320 480 640 0 800 Cell Number DNA content Figure 5.5. Sphinganine increases the number of A0 (sub-GOIG,) cells, indicative of apoptosis, and arrests cell cycle at Gle phase in tumorigenic type I HBEC (one representative histogram). Sub—confluent cells were cultured in the absence (control) or presence of sphinganine at 8 pM for 24 hours and DNA were stained with propidium iodide. The cells in sub-GOIG1 area, indicative of apoptosis, and each cell cycle of 60/6,, 8, and GZIM phases were examined via flow cytometric analysis and calculated using FCS Express version 2.0 and Mod fit cell cycle analysis software as described in Materials and Methods. 142 Tumorigenic 12 . 12- type I HBEC a . a. at 24 h e / s 3 . 3 , o A! lllllll 0245810 SS 383 rrfi 6""C N N L «I O O ‘ G % Cells at 6le % Cells at S % Cells at 60/61 % Apoptotic cells 2o an. 10 1o. Ja‘i‘i‘é‘é 523543513 40 4o so so zob zo 1o- 10 o ........ 0 pM Sphinganine pM Sphingosine Figure 5.6. Sphinganine more potently induces apoptosis of tumorigenic type I HBEC than sphingosine. Both sphingoid bases at high concentrations (8-10 pM) arrest cell cycle at Gle phase with reductions in S and 60/6, phases. Sub-confluent cells were cultured with sphinganine at 0, 3.5, 5, and 8 pM and with sphingosine at 0, 5, 8, and 10 pM for 24 h and DNA were stained with pmpidium iodide. The cells in sub-GOIG1 area, indicative of apoptosis, and each cell cycle of 60/6,, S, and Gle phases were examined via flow cytometric analysis and wiculated using FCS Express version 2.0 and Mod fit cell cycle analysis software as described in Materials and Methods. Where an error bar is not shown, it lies within the dimensions of the symbol. Data are mean 1 SEM (n=2, except n=6 for sphingosine at 0). 143 Sphinganine more potently inhibits the growth and causes death of type I HBE C than sphingosine. To determine the effects of sphinganine on growth and death of type I HBEC with stem cell characteristics in comparison to sphingosine, sub- confluent cells were cultured with sphinganine and sphingosine at various concentrations and culture periods and total nucleic acids were determined as an index of cell number (Figure 5.7-Lefi panel and Figure 5.8-Photographs). Total nucleic acids in control cultures increased more than 3 fold in 6 days, indicating that type I HBEC (Figure 5.7-Lefl panel) grew much slower than tumorigenic type I HBEC (Figure 5.3-Lefi panel). Sphinganine caused concentration- and time- dependent decreases in proliferation of type I HBEC. Specifically, addition of sphinganine at 3.5 and 5 pM reduced total nucleic acids at 3 d by 60 and 90 % and at 6 d by 78 and 94 %, respectively compared to the corresponding controls. Sphinganine at 8 pM completely killed cells. In contrast, sphingosine at 5 pM reduced total nucleic acids at 3 d by 51 % and at 6 d by 74 % (Figure 5.7-Lefi panel and Figure 5.8-Photographs). 144 Total nucleic acids (% of time zero) Figure 5.7. Effects of sphinganine and sphingosine on growth and death of type I HBEC (HME29-A) and type II HBEC (HME29-A). Sub-confluent cells were cultured with sphinganine at 0 (O), 3.5 (V), 5 (V), and 8 (El) pM and with sphingosine at O (O) and 8 (D) pM for 3 and 6 days. Total nucleic acids were measured as an index of cell number. expressed as a percentage of the zero hour. Data are mean 1 SEM (Left panel: n=2, except n=5 for 0 11; Right panef. Data are n=3, except n=6 for 0 d. Means at each culture period with an asterisk (*) are significantly different (P < 0.05) from the corresponding control). Where an error bar is Type I HBEC 400 300 200 1 00 . Sphinganine p 400 300 200 1 00 Sphingosine j l L 4 J J L 0 3 Days Type II HBEC 400 300 200 1 00 _ Sphinganine L414 kg. 1 400 Sphingosine 300 200 ' 100 l I l l l l l 0 3 Days not shown, it lies within the dimensions of the symbol. 145 Results are . Control . Sphinganine 3.5mm Sphinganine 5PM Sphinganine 811M 3 days 6 days Figure 5.8. Sphinganine inhibits the growth and causes deeds of type I HBEC (HME30-4A) (photographs). Sub-confluent cells were cultured with sphinganine at 0, 3.5, 5. and 8 M for 3 and 6 days. Magnification 10X plus 10X. Images in this dissertation are presented in color. 146 Type II HBEC Control ' - Sphinganine -'1 3.5pM Sphinganine 5n" Sphinganine 81M Sphingosine . sum 6 days Figure 5.9. Type II HBEC (HME29-A) are less sensitive to growth inhibitory effects of sphinganine and sphingosine than tumorigenic type I HBEC and type I HBEC at 6 d (photographs). Sub-confluent cells were cultured with sphinganine at 0, 3.5, 5, and 8 pM and with sphingosine at 0 and 8 W for 6 days. Left panel: Areas where the growth of cells are not affected by treatments with sphinganine and sphingosine. Right panel: Amas where the growth of cells are inhibited by treatments with sphinganine and sphingosine. Magnification 10X plus 10X. Images in this dissertation are presented in color. 147 Type II HBE C are less sensitive to growth inhibitory effects of sphinganine and sphingosine than tumorigenic type I HBE C and type I HBE C. To examine the effects of sphingoid bases on grth of type H HBEC (representing normal human breast epithelial cells), sub-confluent cells were cultured with sphinganine and sphingosine at various concentrations and total nucleic acids were determined as an index of cell number (Figure 5.7-Right panel; Figure 5.9-Photographs; Appendix 13: Figure A.14- Photographs). Total nucleic acids in control cultures of type H HBEC increased more than 4 fold in 6 days, indicating that type H HBEC (Figure 5.7-Right panel) grew much slower than tumorigenic type I HBEC (Figure 5.3-Lefi panel) but grew slightly faster than type I HBEC (Figure 5.7-Lefi panel). Sphinganine at 3.5, 5, and 8 pM decreased total nucleic acid concentrations at 3 d by 26, 28, and 61 % and at 6 d by 21, 23, and 53 %, respectively compared to the corresponding controls. Sphingosine at 8 pM reduced total nucleic acids at 3 d by 24 % and at 6 d by 24 % compared to the corresponding control (Figure 5.7-Right panel). These data showed that type H HBEC are less sensitive to growth inhibitory effects of sphingoid bases than tumorigenic type I HBEC (Figure 5.3) and type I HBEC (Figure 5.7-Lefi panel). Sphinganine and sphingosine induce differentiation of type I HBEC to type II HBEC. Type I HBEC were cultured with sphinganine and sphingosine at non- growth inhibitory concentrations for 7 d and the ability of type I HBEC to differentiate to type H HBEC were evaluated by counting morphologically distinguishable colonies of type I, type H, and type I surrounded by type H HBEC (Table 5.1; Figure 5.10- Photographs). 148 Controls (not treated with any chemicals) had ~ 6.1 % differentiation rate and low colony forming efficiency by 7 d. Control vehicles (0.1 % (v/v) of lmM BSA) had ~ 5.4 % differentiation rate. Cells treated with 0.5 % (v/v) of 1 mM BSA increased the differentiation rate to 12.5 % and enhanced colony forming efficiency rate, as shown by the increase in the number of total colonies. This suggests that concentration of BSA higher than that present as the control vehicle might induce the differentiation of type I HBEC to type H HBEC. Sphinganine at 0.05, 0.1, and 0.5 pM increased the differentiation rate to ~ 12.1, 13.3, and 12.4 %, respectively. Sphingosine also increased the differentiation rate to 10.9, 13.5, and 9.8 %, respectively. Cholera toxin (0.1 ng/mL and 1 ng/mL), a positive control known to induce differentiation of type I HBEC to type H HBEC (Kao et al. 1995), increased the number of type H HBEC containing colonies (to 23 and 18 %, respectively for 0.1 and 1 ng/mL), while reduced colony forming efficiency, as shown by the decrease in the number of total colonies. Ethanol at 0.01 and 0.1 % increased the differentiation rate to ~ 13 and 11 %, while reduced colony forming efficiency (Table 5.1). Sphinganine increases the expression of a tumor suppressor protein, maspin during induction of drflerentiation of type I HBE C to type II HBE C. Type I HBEC were cultured with sphinganine at 0.05 and 0.5 pM for 7 d and the expression of a tumor suppressor protein, maspin was determined using immunofluorescence staining (Figure 5.11). Recently our laboratory demonstrated that maspin is expressed in type H HBEC, but not in type I HBEC and in vitro transformed HBEC (immortalized type I I-IBEC, weakly tumorigenic type I, highly tumorigenic type I HBEC) (Saitoh and Chang, 149 unpublished data). Therefore, maspin expression may serve as a biomarker for differentiation of type I HBEC to type H HBEC. As shown in Figure 5.11, maspin expression is not detected in type I HBEC colonies, whereas type H HBEC containing \ colonies show maspin expression. Type H HBEC were more readily detected in sphinganine treated cells compared to the control (Figure 5.11). 150 8228228 .2 2222 2826mm: 2 25 2 82222... 8228 _ 25 +8228 2 25.2 28§=v_us_5+e+=§ 3228228 2 22222 825mm: a 25 B 82222.. 8228 _ 25 .2 28225212552. 2828228 .2 822 828228 852 a 25 .2 2822:1822. .u 25 2 8222.28 8228 _ 25 22 8228 = 25 .2 eaten—5+5 0.52 a 25 2 822022.. a 25 2 8228 .2 22:2 1.5+: .8228 0mm: 2 25 2 525:: um: Own: a 25 .3 Benzene _ 090 v5 .n 25 A 25 mo momma—co me 528:: Me 8.5 ""133. .m 98 m >3 no 33022 883 228222 2828 22 55 2282 28.2 .28 2 H2 2:82 7sz 828mm 2 828222 2522 25 8228 82 22 ma .2 25 2:22. :22 2: 2 28 N 2 .58» 8.... 28:22 2 228 25 222 22 8522 2 888 28» Oman 2 25: 2: 2 we : a. a 2: 2.3 282m a: as 2 cm a : N2 2. :3 282m 3 3. 3 e m m 5 A822 5285 .828 22 2m 2 8 a 2. ea. 2. 2 <3 e: 2 22 t m S 8 .25: 2 222 2220 28 N2 3 8 2 a 8 2222 .c :22 2280 as S 3. a 2 a 3m 21 no 8288225. 22 a 3. an 2 w a: 2: 3 8288225 2: 2m 2m mm 2 2 3m 21 2:. 8228225 22 3 G R 8 m N: 32 no 828225 m2 3. ed on 2 8 PM 21 3 828525 _2 E e an 2 NN Em :1 no... 222225 «a N am a e 2 5 2mm 2: .8 .828 2225 mare + :3. .952. .22. £255: .276 a: "222 .2858... .85: a 25 2 2883:: owe: a 852 8882.22 882 882 28:22 .3 2.5 151 V 3 l 5"“ ‘- ~_*—~~a "cl—d .-T___T. SPH 0.1 pM: 1type ll colony SPH 0.5 ”M: 1 type II colonies. 1 type I differentiating to type II Figure 5.10. Sphinganine and sphingosine induce differentiation of type I HBEC (HME29-A) to type II HBEC (Representative photographs). Type I HBEC were seeded at 5000 cells per 60 mm dish with or without grids and grown for 2 days and then cultured with sphinganine (SA) and sphingosine (SPH) at 0, 0.05, 0.1, and 0.5 pM for 7 days. Fresh medium with and without chemicals were renewed on day 3 and 5. The number of colonies of type I, type II, and type I surrounded by type II HBEC containing colonies were quantitated after 7 d treatments. Magnification 10X plus 4X. Images in this dissertation are presented in color. 152 Control Type I HBEC Control Type I HBEC SA 0.05 pH Type II HBEC SA 0.5 pm Type II HBEC Figure 5.11. Sphinganine increases the expression of a tumor suppressor protein, maspin during induction of differentiation of type I HBEC (HME29- A) to type II HBEC (photographs). Type I HBEC were seeded at 1250 cells per well in 4-well chamber and grown for 2 days and then cultured with sphinganine (SA) at O. 0.05, and 0.5 uM for 7 days. Fresh medium with and without chemicals were renewed on day 3 and 5. On day 7, cells were immunostained with an antibody against maspin. Left panel: Phase contrast; Right panel: Fluorescence. Magnification 10X plus 20X. Images in this dissertation are presented in color. 153 DISCUSSION Sphingolipids have gained much attention for their potential as chemotherapeutic or chemopreventive agents based on in vitro cancer cell and animal studies. For example, in rodent models, both dietary and synthetic sphingolipids inhibit colon carcinogenesis (Dillehay et al. 1994, Schmelz et a1 1996, 1997, 2000, 2001). In addition, a number of studies have demonstrated that the sphingolipid metabolites ceramide and sphingoid bases inhibit growth and induce apoptosis of cancer cells (Ahn and Schroeder 2002b, for review: Merrill et al. 1997, Vesper et al. 1999). However, no study has compared the anti-proliferative and pro-apoptotic efl’ects of sphingoid bases (sphingosine and sphinganine) on human tumorigenic cells to that of human stem and normal cells. The present study is the first to evaluate the chemotherapeutic and chemopreventive potentials of sphingoid bases using the unique normal human breast epithelial cell culture system (type I human breast epithelial cells (HBEC) as normal stem cells; type II HBEC as normal cells; tumorigenic type I HBEC as cancer cells) (Figure 5.2). Since type I HBEC with stem cell characteristics are more susceptible to neoplastic transformation (Kao et al. 1995, Sun et al. 1999), type I HBEC seem to be the target cells for breast carcinogenesis, while type II HBEC may not be the major target cells for carcinogenesis. A major finding of this study is that both sphingosine and sphinganine potently inhibit the growth and induce apoptosis of tumorigenic type I HBEC. Apoptosis was induced in tumorigenic type I HBEC with sphingoid bases ranging from 5-10 M which is lower than the concentrations of ceramide and sphingoid bases necessary to induce apoptosis in other cancer cells (~10-50 pM). This might suggest that breast cancer cells 154 may be more sensitive to sphingoid bases than cancer cells originating in other tissues. It is noteworthy that many other drugs currently used in cancer chemotherapy selectively kill target cancer cells by inducing apoptosis. For example, tamoxifen, widely used for breast cancer treatment, induced apoptosis in both estrogen receptor positive and negative human breast cancer cells (Frankfurt et a1. 1995, Perry et al. 1995, Gelmann et al. 1996, Mandlekar et al. 2000). Moreover, the anticarcinogenic efficacy of many chemotherapeutic agents is associated with sphingolipid metabolism. For example, the chemotherapeutic effect of tamoxifin is reduced when generation of acylated-sphingoid base is impaired, but is enhanced when the degradation of acylated-sphingoid base is blocked (for review: Senchenkov et al. 2001). To our knowledge, this is the first study to show that sphingoid bases at concentrations-inducing apoptosis arrested cell cycle at Gle with reductions in S and Go/Gl phases in human breast tumorigenic cells. The ability to arrest the cell cycle at G2/M appears to be an important characteristic of cancer chemotherapeutic agents. For example, podophyllotoxin, a drug used clinically for cancer treatment (Aisner and Lee, 1990), arrested cell cycle at G2/M with reductions in S and Go/G1 phases and induction of apoptosis in HT-29 human colon cancer cells (Tseng et al. 2002). Interestingly our data suggest that sphingosine has a threshold (greater than 5 but smaller than 8 M4) to cause accumulations of cells in Gz/M phase. Therefore, the Gz/M arrest may not be a good predictor of the degree of apoptosis because sphingosine at both 8 and 10 1.1M caused similar degrees of cell accumulations at G2/M. However, 10 W sphingosine caused a much greater increase of apoptotic cells than 8 uM sphingosine. Furthermore, 155 sphinganine at lower apoptotic concentrations (3.5 and 5 pM) did not arrest cell cycle at G2/M. Another important finding of this study was that both sphingosine and sphinganine inhibited the growth and caused the death of type I HBEC. Type I HBEC display stem cell characteristics and are more susceptible to ne0plastic transformation, telomerase activation, and immortalization (Kao et al. 1995, Kao et al. 1997, Kang et al. 1998, Sun et al. 1999). Thus, sphingoid bases might reduce target cells for breast carcinogenesis. The present study demonstrated that sphinganine is a more potent apoptotic agent for both normal type I HBEC and tumorigenic type I HBEC than sphingosine. This result is consistent with our previous findings in human colon cancer cells (Ahn and Schroeder 2002b) and suggests that the disparity in potency is maintained across cell types. The reason for the greater apoptotic potency of sphinganine than sphingosone is not clear, but is probably related to the relative sensitivity of the molecular targets of the sphingoid bases and/or the activities of the enzymes responsible for sphingoid base metabolism (Sakakura et al. 1998, Baek et al. 2001, for review: Goswami and Dawson, 2000, & Radin, 2001). From a dietary perspective, most complex sphingolipids naturally present in foods contain sphingosine as the backbone. Another important finding of this study is that sphingoid bases seem to meet the criteria for chemotherapeutic agents. That is, sphingoid bases at concentrations that were growth inhibitory and cytotoxic for normal type I HBEC and tumorigenic type I HBEC (sphinganine at 3.5 to 5 M and sphingosine at 8 uM) showed only minor growth inhibitory effects for type II HBEC. This is consistent with those of Sweeney et a1 (1996) 156 who reported that sphingosine induced apoptosis in simian virus 40-transformed rat mesangial cells, but not in their primary culture counterparts (normal rat mesangial cells). This study is the first to evaluate the chemopreventive potential of sphinganine by comparing the effects of sphingoid bases on type I HBEC to those on type II HBEC. Data indicated that sphingoid bases at non-growth inhibitory concentrations (0.05 to 0.5 pM) induced differentiation of type I HBEC to type II HBEC and no dose-response effect was observed within the rage of sphingoid bases concentrations tested. The mechanisms by which sphingoid bases induced differentiation of type I HBEC remains unknown. Ceramide, the acylated form of sphingosine, was first recognized as a differentiating agent for HL-6O human leukemia cells (Okazaki et al. 1989, Okazaki et al. 1990, Kim et al. 1991). Vitamin D3 (Okazaki et al. 1989) and TNFa (Kim et al. 1991) induced this undifferentiated HL-60 cells to become monocytic/macrophage-like cells by increasing ceramide formation. Also, exogenous ceramide mimicked vitamin D3- or TNFa-induced monocytic differentiation of HL-60 cells (Okazaki et al. 1990, Kim et al. 1991). TNFa- induced differentiation of HL-60 cells caused down-regulation of c-myc and the time required to affect c-myc corresponded to the time required for ceramide formation. This suggests that ceramide induced differentiation by regulating c-myc expression. Further studies of ceramide-induced differentiation have been mostly done in neuronal cells and keratinocytes (epidermal epithelial cells) (for review: Huwiler et al. 2000, Luberti et al. 2002). Alternatively, sphingoid bases might increase cellular cAMP to induce the differentiation since cholera toxin (used as a positive control in the current study), a well- known inducer of cAMP, effectively increased the differentiation rate of type I HBEC to type 11 HBEC (Kao et al. 1995). 157 Interestingly, ethanol at 0.01 and 0.1 % also induced differentiation of type I HBEC to type H HBEC. Roles of ethanol on proliferation and differentiation are not clearly understood. Effects of ethanol on proliferation and differentiation might vary depending on different concentrations of ethanol, different cells from different tissue origins, and different culture conditions. For example, ethanol enhanced neural differentiation of PC12 cells with involvement of protein kinase C (for review: Messing 1993). Ethanol inhibited growth and induced apoptosis of hypothalamic beta-endorphin neurons (De et al. 1994). Ethanol inhibited skeletal muscle cell proliferation and delays its differentiation in cell culture (Garriga et al. 2000). Ethanol at 0.3 % stimulated proliferation of MCF-7 human breast epithelial cells, while ethanol at higher than 0.3 % inhibited the proliferation (Izevbigie et al. 2002). This stimulation of proliferation by ethanol was mediated by elevated activity of extracellular signal-regulated kinase-l/Z (ERKl/ERKZ) (Izevbigie et al. 2002). Ethanol at 10 mM increased proliferation and reduced differentiation of head and neck squamous cell carcinoma cells (Komfehl et al. 1999) The current study is the first to show the detection of maspin expression during sphinganine induced differentiation of type I HBEC to type II HBEC. Maspin (mammary serpin) is a novel tumor suppressor gene and an inhibitor of serine proteases. Maspin was originally isolated from normal breast epithelial cells and its expression was decreased during tumor progression (Maass et al. 2002, Shi et al. 2002, Shi et al. 2003). Recently Saitoh and Chang (unpublished data) demonstrated that maspin is expressed in type H HBEC, but not in type I HBEC and in vitro neoplastically transformed HBEC (immortalized type I HBEC, weakly tumorigenic type I, highly tumorigenic type I 158 HBEC). This suggests that maspin expression can serve as a biomarker for status of differentiation of type I HBEC to type H HBEC. Further studies are needed to elucidate the effects of sphinganine on maspin status during differentiation of type I HBEC. In summary, this is the first study that evaluated chemotherapeutic and chemopreventive potentials of sphinganine and sphingosine using a novel cell culture system of HBEC (type I HBEC with stem cell characteristics, type II HBEC, and transformed tumorigenic type I HBEC). The data support the potential for sphingoid bases to be employed as chemotherapeutic and chemopreventive agents against human breast cancer (Figure 5.12). 159 Type I HBEC (stem cells) Type II HBEC Maspin <-> I ‘ ”35”” H I Differentiation ' :4». Sphinganine " Sphingosine " Apoptosis Arrest at GZIM Tumorigenic type I HBEC Figure 5.12. Chemopreventive properties of sphinganine and sphingosine in type | and type II HBEC and chemotherapeutic effects of sphinganine and sphingosine against tumorigenic type I HBEC and type I HBEC. 160 Acknowledgement We express our thanks to: Dr. Dale R. Romsos for his advice and support; Michelle Komosinski and Mark Hagerty for excellent technical assistance on quantitation of colonies of HBEC; Dr. Louis King for advice on flow cytometry analysis; Drs. James E. Trosko and Brad Upham for providing access to their experimental instruments; and Dr. Won 0. Song for her encouragement. 161 CHAPTER 6. CONCLUSION AND SIGNIFICANCE OF THE STUDY 162 CONCLUSION & SIGNIFICANCE OF THE STUDY This study evaluated chemotherapeutic and chemopreventive roles of four major sphingolipid metabolites (sphingosine, sphinganine, ceramide, and dihydroceramide) in colon and breast cancers. Conclusions and significance of the current study are as follows: Conclusion and significance pertaining to colon cancer Data fi'om HT-29 and HGT-116 human colon cancer epithelial cells suggested that sphingoid bases (sphingosine and sphinganine) and Cz-ceramide might possess a potential as chemotherapeutic agents, based on their abilities of inhibiting proliferation, arrest cell cycle, and induce apoptosis within concentration ranges of 20-50 M (Figure 3.8). I examined the importance of the 4,5-trans double bond present in both sphingosine and Cz-ceramide by comparing the effects of these lipids to those of sphinganine and Cz-dihydroceramide, which lack the 4,5-trans double bond (Figure 3.1). My finding of no effect of Cz-dihydroceramide, and a more potent effect of sphinganine than of the other metabolites to induce apoptosis of HT-29 and HGT-116 cells suggest: I) that the 4,5-trans double bond is necessary for the inhibitory effects of Cz-ceramide, but not for inhibitory effects of sphingoid bases; and 2) the more potent effect of synthetic dihydrosphingomyelin (lacks 4,5-trans double bond) than synthetic and dietary sphingomyelin at reducing aberrant colonic foci in CF -1 mice treated with DMH (Schmelz et al. 1997) might be via conversion to sphinganine and/or sphingosine rather than to dihydroceramide and/or ceramide (Figure 2.2). 163 It is unclear why Cz-dihydroceramide lacks biological effects on growth and death of colon cancer cells. One possible explanation is that Cz-dihydroceramide may exhibit limited conversion to flee sphingoid bases. I found that cells exposed to C2- dihydroceramide over 3 to 48 h showed lower intracellular concentrations of free sphingoid bases than those of cells treated with Cz-ceramide (Appendix C, Figure A.l). Quantitation of intracellular concentrations of ceramide as well as free sphingoid bases after sphingoid bases and ceramides treatments would help to clarify possible roles of the 4,5-trans double bond. Both HT-29 and HGT-116 cells responded similme to effects of sphingoid bases and ceramides on growth, cell cycle, and apoptosis. This suggests that several genes showing different mutation status (APC, p53, B-catenin, ras, COX2) between these two cell lines (Table 2.3) do not appear to play major roles for mediating the anti-proliferative and pro-apoptotic actions of sphingoid bases and ceramide. Sphinganine may induce apoptosis of HT-29 cells by altering the balance between pro-apoptotic and anti-apoptotic signaling pathways through JNK2/JNK1, p3 8, and AKT (Figure 2.3; Figure 2.4). My study indicated that sphinganine at an apoptotic concentration (35uM) caused early activation of JNK2/JNK1 and p38, which may be responsible for inhibiting proliferation and stimulating apoptosis. Sphinganine also caused early inhibition of AKT. AKT activation functions in promoting proliferation and inhibiting apoptosis and its activity is enhanced with cancer progression. Conclusion and significance pertaining to breast cancer My study was the first to systemically evaluate the chemotherapeutic and chemopreventive potentials of sphinganine in comparison to sphingosine using a novel 164 normal human breast epithelial cells (HBEC) culture system (Figure 2.7; Table 2.4). The HBEC were developed from mammary tissues of healthy females (type I HBEC with stem cell characteristics and type II HBEC with basal epithelial cell phenotypes). Since type I HBEC are more susceptible to neoplastic transformation, type I HBEC seem to be the target cells for breast carcinogenesis. Tumorigenic type I HBEC were transformed from type I HBEC by a series of oncogenic treatments (Figure 2.7; Table 2.4). The current study advocates that sphingoid bases seem to meet the criteria for chemotherapeutic agents based on following observations (Figure 5.11): 1) Sphingoid bases inhibited proliferation, arrested cell cycle at Gz/M with reductions in S and Go/G1 phases, and induced apoptosis of tumorigenic type I HBEC, with sphinganine being more potent (IC50 for sphinganine 4 uM; sphingosine 6.4 1.1M); 2) Sphingoid bases inhibited the growth and caused the death of type I HBEC with sphinganine being more potent; and 3) Type II HBEC are less sensitive to growth inhibitory effects of sphingoid bases than tumorigenic type I HBEC and type I HBEC. Sphingoid bases might function as chemopreventive agents since sphingoid bases at non-growth inhibitory concentrations (0.05-0.5uM), increased the differentiation rate of type I HBEC to type II HBEC, as detected with expression of a novel tumor suppressor protein maspin in type H HBEC. The mechanisms by which sphingoid bases induced difi‘erentiation of type I HBEC remain unknown. Future study might determine whether c-myc mRN A and cAMP levels mediate actions of sphingoid bases since cholera toxin used as a positive control (Kao et al. 1995) in the current study is known to increase cAMP and c-myc levels were altered during ceramide-induced differentiation of HL-60 cells (Kim et al. 1991). 165 The present study has raised an interesting question of why sphinganine is more potent than sphingosine in inhibiting the growth and causing the death of tumorigenic type I HBEC and type I HBEC as well as HT-29 and HGT-116 cells. The only structural difference between sphinganine and sphingosine is that sphinganine lacks the 4,5-trans double bond (Figure 3.1). Up to now, it is unclear why sphinganine is more potent than sphingosine and ceramide and why 4,5-trans double bond is responsible for the different effect of ceramide and sphingoid bases. Further metabolic studies of ceramide and sphingoid bases including determinations of cellular concentrations of ceramide and sphingoid bases and of activities of enzymes responsible for metabolizing ceramides and sphingoid bases might answer this question. However, fi'om a practical perspective most complex sphingolipids present in foods contain the 4,5-trans double bond. Considerations should be made to compare the efficacy of synthetic sphingoid bases and dietary sphingoid bases to achieve effective doses in vivo. It still remains unclear whether colonic and/or breast tissues are normally exposed to the concentrations of sphingoid bases and/or ceramide used in this study. To use sphingoid bases clinically, further studies are needed to determine digestion, absorption, and excretion of dietary and synthetic sphingolipids in vivo. In the current study, the apoptotic concentration range of ceramide and sphingoid bases for HT-29 and HCT-l 16 human colon cancer cells were 20-50 M and for tumorigenic type I HBEC sphingoid bases at 3.5-10 uM induced apoptosis. Most previous studies reported about 10-50 M as apoptotic concentrations of ceramide and sphingosine, suggesting that breast tumorigenic cells might be more sensitive to sphingoid bases than to cancer cells originated from other tissues. 166 From a clinical view, individual variations in sensitivity of normal breast cells to sphingoid bases could not be excluded. I examined type I and type II HBEC developed from three Caucasian young healthy females (23-26 years old). Here I found a small growth inhibitory effects of sphingoid bases on type II HBEC, however Yang et al (Submitted) did not observe any effect of sphingosine on growth of type II HBEC, suggesting that different individuals might respond differently to sphingoid bases chemotherapy for breast cancer. For future study, it is possible to study the effects of sphingoid bases on the development of human mammary gland in vitro system since type I HBEC together with type II HBEC are able to form terminal end-bud-like structures on Matrigel. Overall, the current study contributes to a better understanding of chemotherapeutic and chemopreventive roles for the sphingolipid metabolites sphingosine, sphinganine, ceramide, and dihydroceramide in colon and breast cancers. My study suggests that sphingoid bases and ceramide might possess chemotherapeutic property against colon cancer, with sphinganine being the most potent. Early activation of JNK and p38 and early inhibition of AKT might mediate sphinganine-induced apoptosis of colon cancer cells. Moreover, the data from HBEC support the potential for sphingoid bases to be employed as chemotherapeutic and chemopreventive agents against human breast cancer. 167 APPENDIX 168 APPENDIX A Table A.l. Mobility of sphingolipids on Thin-Layer Chromatography. Rfvalues Solvent system (v/v) Cer SM SO SPC Gm PhfloSO Ch/Me/Ace/W ND 0.50 0.76 0.19 0.23 0.86 (25:15:43.5) Ch/Me/0.22 % CaClz ND 0.34 0.33 0.11 0.21 ND (60:35:8) Ch/Me/TA/P/0.25% KC] ND ND ND ND ND ND (30:9:25:18z6) Ch/Me/Ace/W 0.97 0.12 0.73 0.03 0.03 0.72 (56:30:42) E/Me (99: 1) 0.57 0.00 0.00 0.00 0.00 0.00 Ch/Me/NI‘LOH 0.94 0.15 0.80 0.04 0.00 0.55 (13:7:1) Ch/Hexane/Me 0.63 0.00 0.05 0.00 0.00 0.04 (10:10:3) Ch/Me/2N NH40H 0.84 0.03 0.20 0.00 0.00 0.10 (40: 10: 1) Ch/Me/Formic acid/W 0.97 0.24 0.76 0.09 0.05 0.73 (56:30:42) Abbreviations used are: ND, nondetectable when same concentrations of sphingolipids were applied; Ch, chloroform; Me, methanol; Ace, acetic acid; TA, triethylamine; P, 2- propanol; W, water; E, diethylether; Cer, Ceramide; SM, sphingomyelin; SO, sphingosine; SPC, sphingophosphorylcholine; Gm, ganglioside Gm; PhytoSO, phytosphingosine. Table A.2. Rf values of sphingolipids on 2-Dimemsional Thin-Layer Chromatography. Rf values Solvent system (Viv) Cer SM ISO SPC Gm lst Diethylether/Methanol 0.57 0.00 0.00 0.00 0.00 (99:1) 2nd Chloroform/Methanol/Acet 0.92 0.13 0.73 0.04 0.04 ic acid/Water (56:30:422) 169 APPENDIX 8 METHODS Quantification of free sphingoid bases. Mass measurements of long-chain (sphingoid) bases were conducted as previously described (Merrill et al. 1988, Riley et al. 1994). Briefly, Czo-sphingosine was added as an internal standard only to samples for the purpose of measuring intracellular fiee sphingoid bases in HGT-116 cells. The sphingoid bases were then extracted with chloroform and methanol and treated with base to remove interfering glycerolipids. After preparation of the o-phthalaldehyde derivatives, the long- chain bases were separated by reverse-phase high-performance liquid chromatography (l-IPLC) using a C13 column eluted isocratically with methanol, 5 mM potassium phosphate, pH 7.0 (91 :9, v/v). The concentrations of sphingoid bases were determined by reference to the Czo-sphingosine, internal standard. Analysis of intemucleosomal DNA fragmentation by gel elctrophoresis. The fragmented DNA was detected using the Genomix cells small scale kit purchased from Talent. The procedure was based on Goruppi et al. (1994). After treatment of cells with sphingoid bases and ceramides for 12 or 24 h, cells were harvested in 150 uL of phosphate buffered saline (PBS). Cells were lysed and aqueous phase was separated using chloroform, acidfication solution, and gel barriers. Cells were precipitated and centrifuged and liquid phase was removed. The DNA pellet was resuspended in distilled water followed by electrophoresis in a 2% agarose gel at 100 V for 70~90 min. The DNA bands stained with ethidium bromide was photographed under UV light. Quantitation of cellular protein using Lowry method. Cellular protein was determined using Lowry method as described (Lowry et al. 1951). The scrape loading/dye transfer assay. Effects of sphingoid bases on gap junctional intercellular communications were determined by the scrape loading/dye transfer assay as described (El-Fouly et al. 1987 and Trosko et al. 2000). Cells were grown until confluency in 35 mm dish and treated with sphingoid bases at various concentrations for 24 and 48 h. Cells were rinsed twice with CaMg-PBS [137 mM NaCl (8g/L), 2.68 mM KCl (0.2g/L), 8.1 mM NazHPO4 (1.15g/L), 1.47 nM KHzPO4 (0.2g/L), 0.68 mM CaClz, 0.49 mM MgClz]. The Lucifer yellow reagent [0.5 mg/mL Lucifer yellow CH lithium salt (Molecular Probes, Eugene, OR), 0.5 mg/mL rhodamine-dextran (Molecular Probes) in CaMg-PBS] was added to cell monolayer and the cell monolayer was wounded in a straight line with a sterile scalpel blade. After incubation of the wounded monolayer in dark room for 3 min, Lucifer yellow was removed fi'om cells and cells were rinsed four times with CaMg-PBS and 4 % (v/v) formalin in CaMg-PBS was added to cells. The distance of dye (Lucifer yellow) transfer from the cells along the scrape line into neighbor cells were observed under fluorescence microscope. 170 APPENDIX c HCT-1 1 6 goells - Dihydroceramide r so Ceramide so 40 20 20 Free sphingoid bases (nmollmg protein) 8 012243648 0012243648 Hours Hours Figure A.1. Effects of exogenous Cz—ceramide and Cz-dihydroceramide on intracellular concentrations of free sphingoid bases in HGT-116 human colon cancer cells. Subconfluent cells were cultured with Cz-ceramide and C2- dihydroceramide at 0 (O), 20 (A), and 50 (0) p.M for 3, 12. 24, or 48 h and cellular concentrations of free sphingoid bases (sphingosine and sphinganine) were quantitated via HPLC. Data represent mean 1 SEM (n=4). Where an error bar is not shown, it lies within the dimensions of the symbol. HGT-116 cells Control 48h SA 20 pM, 48h SA 35 pM, 48h Figure A.2. Effects of sphinganine (SA) on gap junctional intercellular communication on HGT-116 human colon cancer cells (photographs). Confluent cells were cultured with sphinganine at 0, 20. and 35 uM for 48 h. Top panel: Florescence; Bottom panel: Phase contrast. Magnification 10X plus 10X. Images in this dissertation are presented in color. 171 APPENDIX D HT-29 cells :=.- . . ' Control 24h ._ Sphinganine 20 pM, 24h Sphinganine 35 pM, 24h Control 48h Sphinganine _ 20 pM, 48h ‘ r1 Sphinganine r: 35 pM, 48h Figure A.3. junctional intercellular communication on HT—29 human colon cancer cells (photographs). Confluent cells were cultured with sphinganine at 0, 20. and 35 uM for 24 and 48 h. Left panel: Florescence; Right panel: Phase contrast. Magnification 10X plus 20X. images in this dissertation are presented in color. 172 APPENDIX E HT-29 cells Cont SA M Cont Cer M Cont Dh-cer M Figure A.4. Effects of sphinganine, Cz-ceramide, and Cz-dihydroceramide on DNA fragmentation, indicative apoptosis, in HT—29 human colon cancer cells. Subconfluent HT—29 cells were cultured without sphingolipids (control, cont) or with sphinganine (SA) at 10 M for 12h and Cz-ceramide (Cer) and Cz-dihydroceramide (Dh—cer) at 20 uM for 24h. DNA was extracted and analyzed using gel electrophoresis on a 2 % agarose gel at 100V. Abbreviation used: M, DNA marker (123 bp). 173 APPENDIX F HGT-116 cells Cont SD M Cont SA M Hi Cont Cer M Cont Dh-cer M Figure A.5. Effects of sphingosine, sphinganine, Cz-ceramide, and C2- dihydroceramide on DNA fragmentation, indicative apoptosis, in HCT- 116 human colon mncer cells. Subconfluent HOT-116 cells were cultured without sphingolipids (control, cont) or with sphingosine (SO) at 40 l1 M for 24h, sphinganine (SA) at 10 uM for 12h, Cz-ceramide (Cer) and Cz-dihydroceramide (Dh-cer) at 20 uM for 24h. DNA was extracted and analyzed using gel electrophoresis on a 2 % agarose gel at 100V. Abbreviation used: M, DNA marker (123 bp). 174 APPENDIX G a HGT-1 1 6 cells =3 1 g 12 :3: 9 a o O. < 0 .\° Cont 80 SA Figure A.6. Effects of sphingoid bases on apoptotic cell number in HCT- 116 human colon cancer cells. Subconfluent cells were cultured in the absence (control, Cont) or presence of sphingosine (SO) and sphinganine (SA) at 35 uM for 12 h. DNA was then stained with propidium iodide, the cell cycle was examined via flow cytometric analysis, and the percentage of cells in the A0 (sub-Go/G1) region was estimated with the FCS express version 1.0 software. Data are mean 1 SEM (n=2). HGT-116 cells E 100 :3: 75 E": 50 $9.. 25 o 0 Cont 80 SA Figure A.7. Effects of sphingoid bases on cell cycle distribution of HOT-116 human colon cancer cells. Subconfluent cells were cultured in the absence (control, Cont) or presence of sphingosine (SO) and sphinganine (SA) at 35 uM for 12 h. DNA was then stained with propidium iodide, the cell cycle was examined via flow cytometric analysis, and the percentage of cells in each stage of the cell cycle, including GalG1 phase, /4; S phase, D; and GZIM phase, I, was determined using the Multi—cycle DNA content & cell cycle analysis software. The A0 (sub-6016,) cell population was not included in calculation of cell population. Data are mean (n=2). 175 APPENDIX H HGT-116 cells A 125 ‘5. e 100 - 13 . a 15 - ‘I' ' 0 75 - E 50 _ 56 _ f ”r as SA 0 ' 0 : ‘ °\° 13° ' 151 - V _ 125 , '3 90 10% CA '5 3' : 75) < _ s- _2 3. M as: W7 0 o k ‘ 0 a l a L _5 ‘5‘ 166 g 126 126 - _ 106 - 5 ’° 0 6° w " so SA+W7 w SA+W7 0 . . . . . o 3 6 s 12 ' o s a 9 12 hours hours Figure A.8. Effects of a calmodulin antagonist W7 on growth inhibitory effects of sphinganine (SA) in HOT-116 human colon cancer cells. Lelt panel. Subconfluent HGT-116 cells were cultured with SA at 0 (O), 35 uM (O), with W7 at 0 (e), 1 (O), 3 (V), 10 uM (0) and simultaneously with both sphinganine and W7 at control (0), 35 pM SA + 1 uM W7 (0), 35 M SA + 3 M (V), 35 uM SA + 10 pM W7 (0) for 0, 3, and 12 h. Right panel: Subconfluent HGT-116 cells were cultured with SA at 0 (IO), 10 (O), 15 uM (V), with W7 at 0 (e), 1 (O), 3 (V), 10 M (O) and simultaneously with both sphinganine and W7 at control (C), 10 uM SA +1 pM W7 (0), 1O uM SA + 3 uM (V), 10 uM SA +10 uM W7 (6), 15 pM SA +1 pM W7 (Cl), 15 uM SA + 3 uM (I), 15 uM SA +10 pM W7 (I) for 0, 3, and 12 h. Total nucleic acids were determined as an index of cell number. Data represent mean 1 SEM (n=3). Where an error bar is not shown, it lies within the dimensions of the symbol. 176 APPENDIX I RESULTS Efl‘ects of a calmodulin antagonist W7 on growth inhibiton ejfects of sphinganine in HGT-116 human colon cancer cells. HGT-116 cells were treated with a calmodulin antagonist W7 at 1, 3, 5, 10, and 20 M to find the non-growth inhibitory concentration range of W7. Data indicated that W7 at 1, 3, and lOuM did not affect the growth of HOT-116 cells (Figure A.8). HOT-116 cells were treated simultaneously with sphinganine at 35 M and W7 at l, 3, and 10 M for O, 3, and 12 h. The result showed that a calmodulin antagonist W7 did not attenuate the growth inhibitory effect of sphinganine (Figure A.8). However we speculated that this concentration of sphinganine at 35 M was too high and cells were overwhelmed by sphinganine growth inhibitory effect (Figure A.8). As a follow-up experiment, cells were simultaneously treated with sphinganine at lower concentrations of 10 and 15 11M and W7 at 1, 3, 10 M for 0, 3, and 12 h and the similar result was observed (Figure A.8). This finding suggests that the growth inhibitory effect of sphinganine may not be related to an action of calmodulin in HOT-116 human colon cancer cells. 177 APPENDIX J Tumorigenic type I HBEC Control Sphinganine 3.5pM Sphinganine 5PM Sphingosine 5PM Sphingosine BuM Figure A.9. Effects of sphinganine and sphingosine on gap junctional intercellular communication on tumorigenic type I HBEC at 24 h (photographs). Confluent cells were culturedwithsphinganine etc, 3. 5, and 5M and with sphingosine at 0, 5, and 8 uM for 24 h. Left panel. Florescence; Right panel Phase contrast. Magnification 10X plus 10X. Images in this dissertation are presented in color. APPENDIX K Tumorigenic type I HBEC SA 3.5 uM SA 5 uM Sphingosine 5 pM Figure A.10. Sphinganine (SA) more potently inhibits the growth and causes death of tumorigenic type I HBEC than sphingosine at 2 d (photographs). Sub—confluent cells were cultured with sphinganine at 0, 0.5, 2, 3.5, and 5 uM and with sphingosine at 0 and 5 uM for 2 days. 100x magnification. Images in this dissertation are presented in color. Tumorigenic type I HBEC pm 0 1.; 5 10 SA SO Figure A.11. Effects of sphingosine and sphinganine on cell cycle distribution in tumorigenic type | HBEC at 12h. Sub-confluent cells were cultured with sphinganine (SA) at 0 and 3.5 uM and with sphingosine ($0) at O, 5, and 10 uM for 12 h and DNA were stained with propidium iodide. The cells in sub- Go/G1 area, indicative of apoptosis, and the cell cycle were examined via flow cytometric analysis. The percentage of cells in each cell cycle of GolG1 phase, 2; S phase, El; and GZIM phase, I, was determined using the Mod fit cell cycle analysis software. The A0 (sub-6016,) cell population was not included in calculation of cell population. Data are mean 1 SEM (n=2 ). 179 APPENDIX L .C a Tumorigenic type I HBEC 3 Type II HBEC a 73450 g ‘5 300 8 0120 E a T; g 90 g o 200 o g 50 2 -,§. 100 s '5 30 5 *5 Es ° 3 -\° ° _ .. o 0.5 1 5 g .. 0 0-8 g BSA (%) ,2 BSA (%1 Figure A.12. Effects of bovine serum albumin (BSA) on growth of tumorigenic type I HBEC and type II HBEC. Sub-confluent cells were cultured with bovine serum albumin at 0, 0.5, 1, and 5% for 24 hours in tumorigenic type I HBEC and at 0 and 0.8% for 3 days in type II HBEC. Total nucleic acids were measured as an index of cell number. Results are expressed as a percentage of the zero hour. Data Shown are mean 1 SEM (n=3). Where an error bar is not shown, it lies within the dimensions of the symbol. . Fibbtsrolasv * Third type cells Figure A.13. Fibroblasts and the third type of cells are found in human breast epithelial cells (HME23A, HME29A, HME30-4A, HME31-L2A) (photographs). Top panel. 40X magnification. Bottom panel. 100x magnification. Images in this dissertation are presented in color 180 IIIIIIIIII‘ I| APPENDIX M Type II HBEC Control I ‘ Sphinganine ‘ 3.5pM Sphinganine 5PM 3 days Figure A.14. Type II HBEC (HME29A) are less sensitive to growth inhibitory effects of sphinganine and sphingosine than tumorigenic type | HBEC and type I HBEC at 3 d (photographs). Sub-confluent cells were cultured with sphinganine at 0, 3.5, 5,, and 8 uM and with sphingosine at 0 and 8 uM for 3 days. Magnification 100x. Images in this dissertation are presented in color 181 REFERENCES Aaltonen, L., Peltomaki, P., Leach, F., Sistonen, P., Pylkkanen, L., Mecklin, J. P., Jarvinen, H., Powell, 8., Jen, J., Hamilton, 8., Peterson, 6., 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. 50:3S-23S. Ahmad, N., Feyes, D.K., Neiminen, A-L., Agarwal, R., Mukhtar, H. (1997) Green tea constituent epigallocatechin 3-gallate and induction of apoptosis and cell cycle arrest in human carcinoma cells. J. Natl. Caner Inst. 89:1881-1886. Ahn, E. H. & Schroeder, J. J. (2002a) Bioactive sphingolipids are constituents of milk and soy products. J. Food Sci. 67:522-524. Ahn, E. H. & Schroeder, J. J. (2002b) Sphingoid bases and ceramide induce apoptosis in HT-29 and HGT-116 human colon cancer cells. Exp. Biol. Med. 227:345-353. Aisner, J., & Lee, E. J. (1991) Etoposide. Current and future status. Cancer. 67 (Suppl.l): 215-219. 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: Cancer Facts and Figures (1999) Atlanta, GA: American Cancer Society. American Type Culture Collection (ATCC) (2001) ATCC cell lines and hybridomas. Rockville, MI). Armstrong, 8., 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. Asai, A., Kiyozuka, Y., Yoshida, R., Fujii, T., Hioki, K. & Tsubura, A. (1998) Telomere length, telomerase activity and telomerase RNA expression in human esophageal cancer cells: correlation with cell proliferation, differentiation and chemosensitivity to anticancer drugs. Anticancer Res. 18: 1465-1472. Atkins, C. M., Selcher, J. C., Petraitis, J. J., Trzaskos, J. M., Sweatt, J. D. (1998) The MAPK cascade is required for mammalian associative learning. Nature Neurosci 1,602- 609. 182 Autexier C, Greider CW. (1996) Telomerase and cancer: Revisiting the telomere hypothesis. Trends Biochem Sci 21 :387—391 . Awad, A. 3., Von Holtz, V., Jessie, P. C., Carol, S. 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. Awad, A.B., Chen, Y.-C., Fink, C.S. & Hennessey, T. (1996) B—sitosterol inhibits HT -29 human colon cancer cell growth and alters membrane lipids. Back, M.-Y., Yoo, H.-S., Nakaya, K., Moon, D.-C., & Lee, Y.-M. (2001) Sphingolipid metabolic changes during chiral Cz-ceramides induced apoptosis in hmnan leukemia cells. Arch. Pharmacal. Res. 24: 144-149. Ballou, L. R., Chao, C. P., Holness, M. A., Barker, S. C., Raghow, R. (1992) Interleukin- 1-mediated PGE-2 production and sphingomyelin metabolism: evidence for the regulation of cyclooxygenase gene expression by sphingosine and ceramide. J. Biol. Chem. 267: 20044-20050. 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., F elton, J .S., Sato, S.(eds) Wiley-Liss, New York. Bastarrachea, J., Hortobagyi, G. N., Smith, T. L., Lau, S. W., and Buzdar, A. U. (1994) Obesity as an adverse prognostic factor for patients receiving adjuvant chemotherapy for breast cancer. Ann. Intern. Med. 120: 18-25. Batra, S, & Alenfall, J. (1991) Effect of diverse categories of drugs on human colon tumor cell proliferation Anticancer Res. 11:1221-1224. Belair, C. D., Yeager, T. R., Lopez, P. M., Reznikoff, C. A. (1997) Telomerase activity: A biomarker of cell proliferation, not malignant transformation. Proc. Natl. Acad. Sci. USA. 94: 13677—13682. Bhattacharyya, N. P., Ganesh, A., Phear, G., Richards, B., Skandalis, A., & Meuth, M. (1995) Molecular analysis of mutations in mutator colorectal carcinoma cell lines. Hum. Mol. Genet. 4: 2057-2064 Bianchi Salvadori, B. (1986) Intestinal microflora: the role of yogmt in the equilibrium of the gut ecosystem. Int. J. Immunol. 2: 98-] 8S. Bielawaska, A., Crane, H. M., Liotta, D. C., Obeid, L. M. & Hanmm, Y. A. (1993) Selectivity of ceramide-mediated biology: Lack of activity of erythro—dihydrocermiade. J. Biol. Chem. 268: 26226-26232. 183 Biffi A., Coradini D., Larsen R., Riva L., and Fronzo G. D. (1997) Antiproliferative effect of fermented milk on the growth of a human breast cancer cell line. Nutrition and cancer 28: 93-99. Bottomley RH, Condit PT (1968) Cancer families. Cancer Bull 20: 22-24. Bradford, M. M. (1976) A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal. Biochem. 72:248-254. Brattain, M. G., Fine, W. D., Khaled, F. M., Thompson, J., Brattain, D. E. (1981) Heterogeneity of malignant cells fi'om a human colonic carcinoma. Cancer Research 41:1751-1756. Brugarolas J, Jacks T (1997) Double indemnity: p53, BRCA and cancer - p53 mutation partially rescues developmental arrest in Brcal and Brca2 null mice, suggesting a role for familial breast cancer genes in DNA damage repair. Nature Medicine 3(7): 721-722. Brugg, B., Michel, P. P., Agid, Y. & Ruberg, M. (1996) Ceramide induces apoptosis in cultured mesencephalic neurons. J. Neurochem. 66: 733-739. Bryan TM, Englezou A, Dalla-Pozza L, Dunham MA, Reddel RR. (1997) Evidence for an alternative mechanism for maintaining telomere length in human tumors and tumor- derived cell lines. Nat Med 3:1271—1274. 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. Burt, R. W., Petersen GM (1996) Familial colorectal cancer: diagnosis and management. In: Young GP, Rozen P, Levin B, eds.: Prevention and Early Detection of Colorectal Cancer. London, England: WB Saunders, pp 171-194. Burton, K. (1968) Determination of DNA content with diphenylamine. Methods in Enzymology. 12: 163-166. Cai, Z., Bettaieb, A., Mahdani, N. E., Legres, L. G., Stancou, R, Maslliah, J ., & Chouaib, S. (1997) Alteration of the sphingomylein/ceramide pathway is associated with resistance of human breast carcinoma MCF7 cells to tumor necrosis factor-mediated cytotoxicity. J. Biol. Chem. 272: 6918-6926. 184 Cairns, J. (1975) Mutation selection and the natmal history of cancer. Nature 255: 197- 200. Camps, M., Nichols, A., Arkinstall, S. (1999) Dual specificity phosphatases: a gene family for control of MAP kinase fimction. FASEB J. 14:6-16. Cassandra D. Belair, Thomas R. Yeager, Patricia M. Lopez, and Catherine A. Reznikoff. (1997) Telomerase activity: A biomarker of cell proliferation, not malignant transformation. Proc. Natl. Acad. Sci. USA Vol. 94, pp. 13677-13682. Chafouleas, JG, Bolton WE, Hidaka, H, Boyd, AE, Means, AR (1982) Calmodulin and the cell cycle: Involvement in regulation of cell cycle progression. Cell. 18:41-50. Chan, T. A., Morin, P.J., Volgestein, B., Kinzler, K.W. (1998) Mechanisms underlying nonsteroidal anti-inflammatory drug-mediated apoptosis. Proc. Natl. Acad. Sci. 95:681- 686. Chang, C.-C., Boezi, J. A., Warren, S. T., Sabourin, C. L. K., Liu, P. K., Glatzer, L., & Trosko, J. E. (1981) Isolation and characterization of a UV-sensitive hypermutable aphidicolin-resistant Chinese hamseter cell line. Somatic Cell Genetics. 7: 235-253. Chang, C.-C., Sun, W., Cruz, A., Satoh, M., Tai, M.-H., & Trosko, J .E. (2001a) A human breast epithelial cell type with stem cell characteristics as target cells for carcinogenesis. Radiation Res. 155: 201-207. Chang, H. C., Tsai, L. H., Chuang, L. Y., Hung, W. C. (2001b) Role of AKT kinase in sphingosine-induced apoptosis in human hepatoma cells. J. Cellular Physiology. 188: 188-193. Cheng, Y.,Tauschel, H.D., Nilsson, A. & Duan, RD. (1999) Ursodeoxycholic acid increases the activities of alkaline sphingomyelinase and caspase-3 in the rat colon. Scandinavian Journal of Gastroenterology. 34: 915-920. Chiu CP, Harley CB. (1997) Replicative senescence and cell immortality: The role of telomeres and telomerase. Proc Soc Exp Biol Med 214: 99-106. Chmura, S.J., Mauceri, H.J., Advani, S., Heimann, R., Beckett, M.A., Nodzenski, W., Quintans, J ., Kufe, D.W., Weichselbaum, RR (1997) Decreasing the apoptotic threshold of tumor cells through protein kinase C inhibition and sphingomyelinase activation increases tumor killing by ionizing radiation. Cancer Res. 57: 4340-4347. Choi, S. Y., Kahyo, H. (1991) Efl‘ect of cigarette smoking and alcohol consumption in the etiology of cancer of the digestive tract. Int. J. Cancer. 49: 381-3 86. 185 Clark, A. S., West, K., Streicher, S., & Dennis, P. A. (2002) Constitutive and inducible AKT activity promotes resistance to chemotherapy, Trastuzumab, or Tamoxifen in breast cancer cells. Molecular Cancer Therapeutics. 1: 707-717. Clavel-Chapelon, F., Niravong, M., & Joseph, R. R. (1997) Diet and breast cancer: review of the epidemiologic literature. Cancer Detect. Prev. 21: 426-440. Cleary, M. P. , Maihle, N. J. (1997) The role of body mass index in the relative risk of developing premenopausal versus postmenopausal breast cancer. Proc. Soc. Exp. Biol. Med. 216: 28-43. Cohen, L. A., Choi, K., ,Weisburger, J. H., & Rose, D. P. (1986) Effect of varying proportions of dietary fat on the deveIOpment of N-nitrosomethylurea-induced rat mammary tumors. Anticancer Res. 6: 215-218. Cohen, P. (1997) The search for physiological substrates of MAP and SAP kinases in mammalian cells. Trends Cell Biol 7,353-361 Colditz GA, Rosner BA, Speizer FE (1996) Risk factors for breast cancer according to family history of breast cancer. J. of the National Cancer Institute 88(6): 3.65-371. Colditz GA, Willett WC, Hunter DJ, Stampfer MJ, Manson JE, Hennekens CH, Rosner BA (1993) Family history, age, and risk of breast cancer. JAMA: Journal of the American Medical Association 270(3): 338-343. Collaborative Group on Hormonal Factors in Breast Cancer (1997) Breast cancer and hormone replacement therapy: collaborative reanalysis of data from 51 epidemiological studies of 52,705 women with breast cancer and 108,411 women without breast cancer. Lancet 350: 1047-1059. Collins N, McManus R, Wooster R, Mangion, J. Seal, S., Lakhani, S. R., Ormiston, W., Daly, P. A., Ford, D., Easton, D. F., Stratton, M. R. (1995) Consistent loss of the wild type allele in breast cancers fi'om a family linked to the BRCA2 gene on chromosome l3q12-13. Oncogene 10(8): 1673-1675. Comm. Diet Health (1989) Diet and Health-Implications for Reducing Chronic Disease Risk. Washington, DC: Natl. Res. Counc., Natl. Acad. Comm. Diet Nutr. Cancer (1982) Diet, Nutrition and Cancer. Washington, DC: Natl. Res. Counc., Natl. Acad. Counter CM, Hirte HW, Bacchetti S, Harley CB. (1994) Telomerase activity in human ovarian carcinoma. Proc Natl Acad Sci U S A 91:2900—2904. 186 Cowley, S., Paterson, H., Kemp, R, Marshall, C. J. (1994) Activation of MAP kinase kinase is necessary and sufficient for PC 12 differentiation and for transformation of NIH 3T3 cells. Cell 77,841-852. Cummings, J. (1983) Fermentation in the human large intestine: evidence and implications for health. Lancet. 1: 1206-1209. Darzynkiewicz Z., Traganos F., Melamed MR. (1980) New cell cycle compamnents identified by multiparameter flow cytometry. Cytometry 1: 98-108. Davis, D.L., Bradlow, H.L., Wolff, M., Woodruff, T., Hoe], D.G. & Anton, CH. (1993) Medical hypothesis: xenoestrogens as preventable causes of breast cancer. Environ.Health Perspect. 101: 372-377. Davis, M.A., Flaws, J.A., Young, M., Collins, K., Colburn, NH. (2000). Effect of ceramide on intracellular glutathione determines apoptotic or necrotic cell death of JB6 tumor cells. Toxicological Sciences. 53: 48-55. Dbaibo, G.S., Pushkareva, M.Y., Rachid, R.A., Alter, N., Smyth, M.J., Obeid, L.M., & Hannun, Y.A. (1998) p53-dependent ceramide response to genotoxic response. J. Clin. Invest. 102-329—339. De, A., Boyadjieva, N. I., Pastorcic, M., Reddy, B. V., & Sarkar, D. K. (1994) Cyclic AMP and ethanol interact to control apoptosis and differentiation in hypothalamic beta- endorphin neurons. J. Biol. Chem. 269: 26697-26705. Del Bino, G. & Darzynkiewicz, Z. (1991) Camptothecin and teniposide, or 4’-(9- acridinylamino)-3-methane-sulfon-m-anisidide, but not mitoxantrone or doxorubucun, induces degradation of nuclear DNA in the S phase of HL-60 cells. Cancer Res. 51: 1165-1169. Dillehay, D. L., Webb, S. K., Schmelz, E. M., Merrill, A. H., Jr. (1994) Dietary sphingomyelin inhibits 1,2-dimethylhydrazine—induced colon cancer in CF] mice. J. Nutr. 124: 615-620. Dong, C., Yang, D.D., Wysk, M., Whitmarch, A.J., Davis, R.J., Flavell, RA. (1998) Defective T cell differentiation in the absence of JNK1. Science. 282:2092-2095. Dressler, K. A., Mathias, S., Kolesnick, R. N. (1992) TNF-ct activates the sphingomyelin signal transduction pathway in a cell-free system. Science. 255: 1715-1718. Dudley, D. T., Pang, L., Decker, S. J., Bridges, A. J., Saltiel, A. R. (1995) A synthetic inhibitor of the mitogen-activated protein kinase cascade. Proc. Natl. Acad. Sci. USA 92,7686-7689. 187 Duncan JL, Kyle J (1982) Family incidence of carcinoma of the colon and rectum in north-east Scotland Gut 23(2): 169-171. Durha,, A. C. H., Walton, J. M. (1982) Calcium ions and the control of proliferation in normal and cancer cells. Biosci. Rep. 2:15-30. Easton DF (1994) Cancer risks in A-T heterozygotes. Int. J. Radiat. Biol. 66 (6 Suppl): S177-82. El-Deiry, W. S., Nelkin, B. D., Celano, P., Yen, R. W., Falco, J. P., Hamilton, S. 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. El-Fouly, M. H., Trosko, J. E., Chang, C.-C. (1987) Scrape-loading and dye transfer: a rapid and simple techinique to study gap junctional intercellular communivation. Exp. Cell. Res. 168: 422-430. 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, - trimethyl sphingosine derivatives on in vitro and in vivo growth of human tumor cells in nude mice. Cancer Res. 51: 1613-1618. Erickson, B.A., Longo, W.E., Panesar, N., Mazuski, J.E. & Kaminski, BL. (1999) The effect of selective cyclooxygenase inhibitors on intestinal epithelial cell mitogenesis. Journal of Surgical Research. 81: 101-107. Esteve, J., Kriker, A., Ferlay, J., & Parkin, D. M. (1993) Facts and figures of cancer in the European community. Lyon: INtmational Agency for Reseach on Cancer. Fanger, G. R., Gerwins, P. Widmann,C., Jarpe, M. B., Johnson,G. L. (1997) MEKKs, GLKs, MLKs, PAKs, TAKs, and Tpls: upstream regulators of the c-Jun amino terminal kinases?. Curr. Opin. Genet. Dev. 7, 67-74. F earon, E., Cho, K., Nigro, J. (1990) Identification of a chromosome 18q gene that is altered in colorectal cancers. Science. 247: 49—56. F earon, E.R., Volgestein, 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. F errell, J. E., Jr (1996) MAP kinases in mitogenesis and development. Curr. Top. Dev. Biol. 33,1-60. 188 Feuer EJ, Wun LM, Boring CC, Flanders WD, Timmel MJ, Tong T (1993) The lifetime risk of developing breast cancer. Journal of the National Cancer. Institute 85(11): 892- 897. Fisher, D.E. (1994) Apoptosis in cancer therapy: crossing the threshold. Cell 78: 539- 542. Flamigni F, Faenza I, Marmiroli S, Stanic' I, Giaccari A, Muscari C, Stefanelli C, & Rossoni C. (1997) Inhibition of the expression of ornithine decarboxylase and c-myc by cell-permeant ceramide in difluoromethylornithine-resistant leukaemia cells. Biochem. J. 324: 783-789. Frankfurt, O. S., Sugarbarker, E. V., Robb, J. A., & Villa, L. (1995) Synergistic induction of apoptosis in breast cancer cells by tamoxifen and calmodulin inhibitors. Cancer Lett. 97: 149-154. Freedman, L. 8., Clifford, C., & Messina, M. (1990) analysis of dietary fat, calories, body weight, and the development of mammary tumors in rats and mice: a review. Cancer Res. 50: 5710-5719. Fuchs CS, Giovannucci EL, Colditz GA, Hunter DJ, Speizer FE, Willett WC (1994) A prospective study of family history and the risk of colorectal cancer. New England J. of Medicine 331(25): 1669-1674. Ganjam, L. S., Thornton, W. H., Jr., Marshall, R. T., MacDonald, R. S. (1997) Antiproliferative effects of yogurt fiactions 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. Garber JE, Goldstein AM, Kantor AF, et al. (1991) Follow-up study of twenty-four families with Li-Fraumeni syndrome. Cancer Res 51 (22): 6094-7. Garriga, J ., Adanero, E., Femandez-Sola, J., Urbano-Marquez, A., & Cusso, R. (2000) Ethanol inhibited skeletal muscle cell proliferation and delays its differentiation in cell culture. Alcohol Alcohol. 35: 236-241. Gelmann E. P. (1996) Tamoxifen induction of apoptosis in estrogen receptor-negative cancers: new tricks for an old dog?. J. Natl. Cancer Inst. 88: 224-226. Gerharsson, M., Norel], S. E., Kiviranta, H., Pedersen, N. L., Ahlbom, A. (1986) Sedentary jobs and colon cancer. Am J. Epidemic]. 123: 775-780. 189 Gill, Z. P., Perks, C. M., Newcomb, P. V., & Holly, J. M. (1997) Insulin-like growth factor-binding protein (IGFBP-3) predisposes breast cancer cells to programmed cell death in a non-IGF-dependent manner. J. Biol. Chem. 272: 25602-25607. Giovannucci, E., & Goldin, B. (1997) The role of fat, fatty acids, and total energy intake in the etiology of human colon cancer. Am. J. Clin. Nutr. 66: 15648-157] S. Glass AG, Hoover RN. (1990) Rising incidence of breast cancer: relationship to stage and receptor status. J. Natl. Cancer Inst. 82:693—96 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 epiderrnoid carcinoma cells: evidence that ceramide may mediate sphingosine action. J. Biol. Chem. 266: 16092-16097. Gorczyca, W., Gong, J ., Ardelt, B., Tragances, F. & Darzynkiewicz, Z. (1993) The cell cycle related differences in susceptibility of HL-60 cells to apoptosis induced by various antitumor agents. Cancer Res. 53: 3186-3192. Goruppi, S., Gustinchin, S., Brancolini, C., Lee, W. M., & Schneider, C. (1994) Dissection of c-myc domains involved in S phase induction of NIT-I3T3 fibroblasts. Oncogene. 9: 1537-1544 Goswami, R., & Dawson, G. (2000) Does ceramide play a role in neuronal cell apoptosis? J. Neuroscience Research. 60: 141-149. Graham, S., Marshall, J ., Haughey, B., Mittelman, A., Swanst M., Zielezny, M., Byers, T., Wilkinson, G., & West, D. (1988) Dietary epidemiology of cancer of the colon in western New York. Am. J. Epidemiol. 128:490—503. Green, DR. (1997) A Myc-induced apoptosis pathway surfaces. Science 278: 1246- 1247. Groden J, Joslyn G, Samowitz W, Jones D, Bhattacharyya N, Spirio L, Thliveris A, Robertson M, Egan S, Meuth M. (1995) Response of colon cancer cell lines to the introduction of APC, a colon-specific tumor suppressor gene. Cancer Res 55:1531-1539. Grubbs CJ, Famel] DR, Hill DL, McDonough KC. (1985) Chemoprevention of N- nitroso-N-methylurea-induced mammary cancers by pretreatment with 17B-estradiol and progesterone. J Natl Cancer Inst. 74:927-31. 190 Gudas JM, Li T, Nguyen H, Jensen D, Rauscher FJ, Cowan KH (1996) Cell cycle regulation of BRCA1 messenger RNA in human breast epithelial cells. Cell Growth and Differentiation 7(6): 717-723. Hakomori, S. (1983) In: Sphingolipids in Biochemistry (Kanfer, J. N., Hakomori, S., ed.), pp. 1, Plenum, New York. Hannun, Y. A. (1997) Apoptosis and the Dilemma of Cancer Chemotherapy. Blood. 89: 1845-1853. Henderson, B. E., Ross, R. K., & Pike, M. C. (1993) Hormonal chemoprevention of cancer in women. Science. 259: 633-638. Hill, M. J. (1971) Fecal steroid composition and its relationship to cancer of the large bowel. J. Pathol. 104: 129-139. Hill, M. J., Morson, B. C., & Bussey, H. J. R. (1978) Aetiology of adenoma-carcinoma sequence in large bowel. Lancet. 1: 245-247. His, L. C., Baek, S. J ., Eling, T. E. (2000) Lack of cyclooxygenase-2 activity in HT-29 human colorectal carcinoma cells. Experimental-Cell-Research. 256 (2): 563-570. Hiyama E., Hiyama K., Yokoyama T., Matsuura Y., Piatyszek MA., Shay JW. (1995) Correlating telomerase activity levels with human neuroblastoma outcomes. Nat Med. 1:249-255. Hiyama K, Hirai Y, Kyoizumi S, Akiyama M, Hiyama E, Piatyszek MA, Shay JW, Ishioka S, Yamakido M. (1995) Activation of telomerase in human lymphocytes and hematopoietic progenitor cells. J Immunol 155:3711—3715. Hiyama K, Ishioka S, Shirotani Y, Inai K, Hiyama E, Murakami I, Isobe T, Inamizu T, Yamakido M. (1995) Alterations in telomeric repeat length in 11mg cancer are associated with loss of heterozygosity in p53 and Rb. Oncogene. 10:937-44. Hiyama, E., L Gollahon, T Kataoka, K Kuroi, T Yokoyama, AF Gazdar, K Hiyama, MA Piatyszek and JW Shay (1996) Telomerase activity in human breast tumors. J. Natl. Cancer Institute. 88:116-122. Hohaus S, Voso MT, Orta-La Barbera E, Cavallo S, Bellacosa A, Rutella S, Rumi C, Genuardi M, Neri G, Leone G. (1997) Telomerase activity in human hematopoietic progenitor cells. Haematologica 82:262-268. Holt, S.E., Shay, J .W. (1999) Role of telomerase in cellular proliferation and cancer. J. Cellular Physiology. 180:10-1 8. 191 Holtz, R.L, Fink, C.S., Awad, AB. (1998) B—sitostero] activates the sphingomyelin cycle and induces apoptosis in LNCaP human prostate cancer cells. Nutrition and Cancer. 32: 8-12. Hoos, A., Hepp, H.H., Kaul, S., Ahlert, T., Bastert, G., Wallwiener, D. (1998) Telomerase activity correlates with tumor aggressiveness and reflects therapy effect in breast cancer. International J. Cancer. 79:8-12. Hovland A, Nahreini P, Andreatta C, Edwards-Prasad J, Prasad K. (2001) Identifying genes involved in regulating differentiation of nemoblastoma cells. J Neurosci Res 64:302—3 10. Hsiao R, Sharma HW, Ramakrishnan S, Keith E, Narayanan R. (1997) Telomerase activity in normal human endothelial cells. Anticancer Res 17:827—832. Huang RFS, Ho YH, Lin HL, Wei J S, Liu, TZ. (1999) Folate deficiency induces a cell cycle—specific apoptosis in HepGZ cells. J Nutr 129: 25-31. Ichijo, H., Nishida, E., Irie, K., Dijke, P. T., Saitoh, M., Moriguchi, T., Takagi, M., Matsumoto, K., Miyazono, K., Gotoh, Y. (1997) Induction of apoptosis by ASK], a mammalian MAPKKK that activates SAPme and p38 signaling pathways. Science 275,90-94. Ide T. (1995) Telomerase activity in human liver tissues: comparison between chronic liver disease and hepatocellular carcinomas. Cancer Res. 55: 2734-6. Ilyas, M., Tomlinson, I. P. M., Rowan, A., Pignatelli, M., Bodmer, W. F. (1997) B- Catenin mutations in cell lines established fi'om human colorectal cancers. Proc. Natl. Acad. Sci. USA. 94:10330-10334. Imaizumi, K., Tominaga, A., Sato, M. & Sugano, M. (1992) Efl‘ects of dietary sphingolipids on levels of serum and liver lipids in rats. Nutr. Res. 12: 543-548. International 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. Izevbigie, E. B., Ekunwe, S. T., Jordan, J ., & Howard, C. B. (2002) Ethanol modulates the growth of human breast cancer cells in vitro. Exp. Biol. Med. 227: 260-265. Jarvis, W. D., Fornari, F. A. Jr., Auer, K. L., Freemerman, A. J., Szabo, E., Birrer, M. J., Johnson, C. R., Barbour, S. E., Dent, P., Grant, S. (1997) Coordinate regulation of stress- and mitogen-activated protein kinases in the apoptotic actions of ceramide and sphingosine. Molecular-Pharmacology. 52:935-947. 192 Jarvis, W. D., Fomari, F. A., Traylor, R. S., Martin, H. A., Kramer, L. B., Erukulla, R. K., Bittrnan, R. & Grant, S. (1996) Induction of apoptosis and potentiation of ceramide- mediated cytotoxicity by sphingoid bases in human myeloid leukemia cells. J. Biol. Chem. 271: 8275-8284. Jonason, A.S., Kunala, S., Price, G.J., Restifo, R.J., Spinelli, H.M., Persing, J.A., Leffell, D.J., Tarone, R.E., Brash, DE. (1996) Frequent clones of p53-mutated keratinocytes in normal human skin. Proc. Natl. Acad. Sci. USA. 93:14025-14029. Jordan, V. C., & Morrow, M. (1999) Tamoxifen, raloxifene, and the prevention of breast cancer. Endocrine Reviews. 20: 253-278. Kaitiyar, S.K., Mukjtar, H. (1996) Tea in chemoprevention of cancer: Epidemiologic and expermintal studies. Int. J. Oncol. 8:221-238. Kampman, 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. Kang, K. S., Morita, I., Cruz, A., Jeon, Y. J., Trosko, J. E., and Chang, C. C. (1997) Expression of estrogen receptors in normal human breast epithelial cell type with luminal and stem cell characteristics and its neoplastically transformed cell lines. Carcinogenesis 18: 251-257. Kang, K. S., Sun, W., Nomata, K, Morita, I, Cruz, A, Liu, CJ, Trosko, JE, Chang, CC (1998) Involvement of tyrosine phosphorylation of p185 (c-erbB2/neu) in tumorigenicity induced by x-rays and the neu oncogene in human breast ephithelial cells. Molecular Carcinogenesis. 21: 225-233. Kang, K.-S., Morita, 1., Cruz, A., Jeon Y. J., Trosko, J. E., and Chang, C.-C. (1997) Expression of estrogen receptors in a normal human breast epithelial cell type with luminal and stem cell characteristics and its neoplastically transformed cell lines. Carcinogenesis, 18: 251- 257. Kannan, S., H Tahara, H Yokozaki, B Mathew, KR Nalinakumari, MK Nair and E Tahara (1997)Telomerase activity in premalignant and malignant lesions of human oral mucosa. Cancer Epidemiology Biomarkers & Prevention. 6:413-420 Kao, C. Y., Nomata, K., Oakley, C. S., Welsch, C. W., and Chang, C. C. (1995) Two types of normal human breast epithelial cells derived fi'om reduction mammoplasty: phenotypic characterization and responses to SV 40 transfection. Carcinogenesis 16:531- 538. Kao, C. Y., Oakley, C. S., Welsch, C. W., Chang, C.-C. (1997) Growth requirements and neoplastic transformation of two types of normal human breast epithelial cells derived 193 from reduction mammoplasty. In Vitro Cellular and Developmental Biology. Animal. 33: 282-288. Karasavvas, N., Erukulla, R K., Bittrnan, 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. Kato, Y., Tapping, R. 1., Huang, S., Watson, M. H., Ulevitch, R. J., Lee, J.-D. (1998) Bmkl/ErkS is required for cell proliferation induced by epidermal growth factor. Nature (London) 395,713-716. Kelsey JL, Hom-Ross PL. (1993) Breast cancer: magnitude of the problem and descriptive epidemiology. Epidemic]. Rev. 15:7—16 Kelsey JL. (1993) Breast cancer epidemiology: summary and future directions. Epidemic]. Rev. 15:256—63 Kim J. H., Oh, S. 0., Jun, S. S., Jung, J. S., Woo, J. S., Kim, Y. K., & Lee, S. H. (1999) Activation of SAPK and increase in Bak levels during ceramide and indomethacin- induced apoptosis in HT29 cells. Korean Journal of Physiology and Pharmacology. 3: 75-82. Kim NW, Piatyszek MA, Prowse KR, Harley CB, West MD, Ho «PL, Coviello GM, Wright WE, Weinrich SL, Shay JW. (1994) Specific association of human telomerase activity with immortal cells and cancer. Science 266:2011—2015. Kim, M. S., Lee, D. Y., Wang, T., Schroeder, J. J. (2001) Fumonisin Bl induces apoptosis in LLC-PKl renal epithelial cells via a sphinganine and calmodulin dependent pathway. Toxicol Appl Pharmacol. 176:118-126. 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 or and y— interferon-Specific role in differentiation. J. Biol. Chem. 266: 484—489. Kimball, John w. Biology. Addison Wesley Publishing Co. 1983. 5‘11 edition. 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. Kirk, K.E., Harmon, B.P., Reichardt, I.K., Sedat, J.W. & Blackburn, EH. (1997) Block in anaphase chromosome separation caused by a telomerase template mutation. Science 75: 1478-1481. Kiyono T, Foster SA, Koop JI, McDougall JK, Galloway DA, Klingelhutz AJ. (1998) Both Rb/p16INK4a inactivation and telomerase activity are required to immortalize human epithelial cells. Nature 396:84—88. 194 Klrufeld, D. M., Lloyd, L. M., Welch, C. B., Davis, M. J., Tulp, O. L., and Kritchevsky, D. (1991) Reduction of enhanced mammary carcinogenesis in LA/N-cp (corpulent) rats by energy restriction. Proc. Soc. Exp. Biol. Med. 196:381-384. Klurfeld, D. M., Weber, M. M., & Kritchevsky, D. (1987) Inhibition of chemically induced mammary and colon tumor promotion by caloric restriction in rats fed increased dietary fat. Cancer Res. 47: 2759-2762. Knowles LM, Milner JA. (1998) Depressed p34cdc2 kinase activity and Gle phase arrest induced by diallyl disulfide in HGT-15 cells. Nutrition and Cancer 30:169-174. Knowles LM, Milner JA. (2001) Possible mechanism by which ally] sulfides suppress neoplastic cell proliferation. J Nutr 13]: 1061S-1066S. Komfehl, J ., Temmel, A., Formanek, M., & Knerer, B. (1999) Effects of ethanol treatment of proliferation and differentiation in a head and neck squamous cell carcinoma cell line. Alcohol. Clin. Exp. Res. 23: 1102-1107. Komhauser, J. M., Greenberg, M. E. (1997) A kinase to remember: dual roles for MAP kinase in long-term memory. Neuron 18,839-842. Ku, W.C., Cheng, A.J. & Wang, TC. (1997) Inhibition of telomerase activity by PKC inhibitors in human nasopharyngeal cancer cells in culture. Biochem. Biophys. Res. Commun. 241: 730-736. Kummer J. L., Rao P. K., & Heidenreich K. A. (1997) Apoptosis induced by withdrawal of trophic factors is mediated by p38 mitogen-activated protein kinase. J. Biol. Chem. 272: 20490—20494. Kutchera W, Jones DA, Matsunarni N, Groden J, McIntyre TM, Zimmerman GA, White RL, Prescott SM. (1996) Prostaglandin H synthase 2 is expressed abnormally in human colon cancer: Evidence for a transcriptional effect. Proc. Natl. Acad. Sci. USA 93:4816- 4820. Kyo S, Kunimi K, Uchibayashi T, Namiki M, Inoue M. (1997) Telomerase activity in human urothelial tumors. Am. J. Clin. Pathol. 107(5):555-560. Lamartiniere CA. (2000) Protection against breast cancer with genistein: a component of soy. Amer. J. Clinical Nutr. 71:1705S-1707S. Lamartiniere CA, Murrill WB, Manzolillo PA, Zhang, J. X., Barnes, S., Zhang, X., Wei, H., Brown, N. M. (1997) Genistein alters the ontogeny of mammary gland development and protects against mammary cancer in rats. Proc Soc Exp Biol Med 1997;217:358—364. 195 Landberg, G., Nielsen, N.H., Nilsson, P., Emdin, S.O., Cajander, J. & Roos, G. (1997) Telomerase activity is associated with cell cycle deregulation in human breast cancer. Cancer Res. 57: 549-554. Lee HP, Gourley L, Duffy SW, Esteve J, Lee J, Day NE. (1991) Dietary effects on breast cancer risk in Singapore. Lancet. 337:1197—200. Lee, I. M., Paffenbarger, R. S., Jr., Hsieh, C. (1991) Physical activity and risk of developing colorectal cancer among college alumni. J. Natl. Cancer Inst. 83: 1324—1329. Lee, M. M., & Lin, S. S. (2000) Dietary fat and breast cancer. Ann. Rev. Nutr. 20:221- 248. Levy, G.N. (1997) Prostaglandin H synthases, nonsteroidal anti-inflammatory drugs, and colon cancer. FASEB J. 11:234-247. Lewin, B.. (1997) Chapter 37. Oncogenes and cancer (subtitle include Irnmortalization and transformation) pp. 1131-1172, In: Genes. 6‘“ edition Oxford University Press, Inc., New York. USA. Li ZR, Hromchak R, Mudipalli A, Bloch A. (1998) Tumor suppressor proteins as regulators of cell differentiation. Cancer Res 58:4282—4287. Li, 1.-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. Lin, Y, H Miyamoto, K Fujinami, H Uemura, M Hosaka, Y Iwasaki and Y Kubota (1996) Telomerase activity in human bladder cancer. Clinical Cancer Research. 2:929- 932. 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: 5 761- 5760. Lipkin, M., Reddy, B., Newmark, H., Lamprecht, S. A. (1999) Dietary factors in human colorectal cancer. Ann. Rev. Nutr. 19: 545-586. Lippman, S.M., Lee, J.J., Sabichi, AL. (1998) Cancer chemoprevention: progress and promise. J. Natl. Cancer Inst. 90:1514-1528. Liu, D., el-Hariry, 1., Karayiannakis, A.J., Wilding, J ., Chinery, R., Kmiot, W., McCrea, P.D., Gullick, W.J., Pignatelli, M. (1997) Phosphorylation of beta-catenin and epidermal growth factor receptor by intestinal trefoil factor. laboratory Investigation. 77:557-563. 196 Lodish, H., Arnold Berk, S. Lawrence Zipursky, Paul Matsudaira. (1999) Molecular Cell Biology, Fourth edition. W.H. Freeman Co., New York, NY. 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. Lorentz, O., Cadoret, A., Duluc, Isabelle, Capeau, J., Gespach, C., Cherqui, G., Freund, J. N. (1999) Downregulation of the colon tumour-suppressor homeobox gene Cdx-2 by oncogenic ras. Oncogene. 18: 87-92. 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. Maass, N., Nagasaki, K., Ziebart, M., Mundhenke, C., & Jonat, W. (2002) Expression and regulation of tumor suprressor gene maspin in breast cancer. Clin. Breast Cancer. 3: 281-287. 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. Malkin D. (1993) The Li-Fraumeni syndrome. Cancer: Principles and Practice of Oncology Updates 7: 1-14. Mandlekar, S., Yu, R., Tan, T. H., Kong, A. N. (2000) Activation of caspase-3 and c-Jun NH2-terminal kinase-1 signaling pathways in tamoxifen-induced apoptosis of human breast cancer cells. Cancer Res. 60: 5995-6000. Mandlekar, S., Yu, R., Tan, T. H., Kong, A. N. (2000) Activation of caspase-3 and c-Jun NH2-terminal kinase-l signaling pathways in tamoxifen-induced apoptosis of human breast cancer cells. Cancer Res. 60: 5995-6000. 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. Mansour, S. J., Matten, W. T., Hermann, A. S., Candia, J. M., Rong, S., Fukasawa, K., Vande Woude, G. F ., Ahn, N. G. (1994) Transformation of mammalian cells by constitutively active MAP kinase kinase. Science 265,966-970. 197 Markert, CL. (1968) Neoplasia: a disease of cell differentiation. Cancer Res. 28: 1908- 1914. Marshall, C. J. (1995) Specificity of receptor tyrosine kinase signaling: transient versus sustained extracellular signal-regulated kinase activation. Cell. 80:179-185. Marshall, B. (1993) Search for a killer: focus shifts from fat to hormones. Science. 259: 618-621. Martin, D., & Lenardo, M. (1994) 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. pp 3.17.1-3.17.39. Mathias, S., Dressler, K. A., Kolesnick, R N. (1991) Characterization of a ceramide- activated protein kinase: stimulation by TNF-ot. Pro. Natl. Acad. Sci. USA. 88: 10009- 10013. Mathias, S., 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. Matroule, J .Y., Bonze, G., Morliere, P., Paillous, N., Santus, R., Bours,V., Piette, J. (1999) Pyropheophorbide-a methyl ester-mediated photosensitization activates transcription factor NF-kappa B through the interleukin-1 receptor-dependent signaling pathway. J. Biol. Chem. 274: 2988-3000. Means, A. R (1988) Molecular mechanisms of action of calmodulin. Recent Prog. Hormone Res. 44: 223-262. . Merrill, A. H. Jr. (1991) Cell regulation by sphingosine and more complex sphingolipids. J. Biomemb. Bioenerget. 23: 83-104. Merrill AH 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(2): 373-381. 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: l677S-1682S. Merrill, A.H., Jr., Schmelz, E.M., Dillehay, D.L., Spiegel, S., Shayman, J.A., Schroeder, J.J., Riley, RT., Voss, K.A. & Wang, E. (1997) Sphingolipids-The enigmatic lipid class: biochemistry, physiology, and pathopyhsiology. Toxicol. Appl. Pharmacol. 142: 208- 225. 198 Messina, M.J., Persky, V., Setchell, K.D. & Barnes, S. (1994) Soy intake and cancer risk: a review ofthe in vitro and in vivo data. Nutr.Cancer 21: 113-131. Michaelson, J. (1991) In: Apoptosis: the molecular basis of cell death (Tomei, L. D., Cope, F. 0., eds) pp. 31-46, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY. Miller BA, Ries LAG, Hankey BF, Kosary CL, Harras A, et al. (1993) SEER Cancer Statistics Review, 1973—1990. NIH Pub]. No. 93—2789. Bethesda, MD: Natl. Cancer Inst. 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. Mukhtar, H, Ahmad, N. (1999) Cancer chemoprevention: future holds in multiple agents. Toxicol. Appl. Pharmacol. 158:207-210. Mutai, M., Tanaka, R. (1987) Ecology of Bifidobacteria in the human intestinal microflora. Bifidobactria Microflora. 6: 33-41. Narayan S, Jaiswal AS. (1997) Activation of Adenomatous Polyposis Coli (APC) Gene Expression by the DNA-alkylating Agent N-Methle-nitro-N-nitrosoguanidine Requires p53. J Biol Chem 272:30619-30622. National Cancer Institute (1999) In: Chemotherapy and you: a guide to self-help during cancer treatment. Bethesda, MD. National Cancer Institute (2002) Genetics of Breast and Ovarian Cancer: cancer genetics- health professionals. CancerNet. www. cancer. gov Negri E., Braga C., La Vecchia, C., Franceschi, S., F iliberti, R., Montella, M., Falcini, F., Conti, E., Talamini, R. (1998) Family history of cancer and risk of colorectal cancer in Italy. British Journal of Cancer. 77: 174-179. 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. Nilsson, A. (1968) Metabolism of sphingomyelin in the intestinal tract of the rat. Biochim Biophys. Acta 164: 575-584. Nilsson, A. (1969) Metabolism of cerebrosides in the intestinal tract of the rat. Biochim Biophys. Acta187: 113-121. 199 Norrback KP, Dahlenborg K, Carlsson R, Roos G. (1996) Telomerase activation in normal B lymphocytes and non-Hodgkin's lymphomas. Blood 88:222—229. 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. Ohta, H., Sweeney, B.A., Masamune, A., Yatomi, Y., Hakomori, S. & Igarashi, Y. (1995) Induction of apoptosis by sphingosine in human leukemic HL-60 cells: a possible endogenous modulator of apoptotic DNA fiagmentation occurring during phorbol ester- induced difl‘erentiation. Cancer Res. 55: 691-697. Okarnoto, S., Sakai, M., Uchida, J. & Saito, H. (1996) 5-fluorouracil induces apoptotic cell death with G2 phase arrest in human breast cancer grated in nude mice. Anticancer Res. 16: 2699-2704. Okayasu, I., Mitomi, H., Yamashita, K., Mikami, T., Fujiwara, M., Kato, M. & Oshimura, M. (1998) Telomerase activity significantly correlates with cell difl‘erentiation, proliferation and lymph node metastasis in colorectal carcinomas. J .Cancer Res.C1in.Oncol. 124: 444-449. 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 la, 25-dihydroxy vitamin Dg-induced HL-60 cell differentiation. J. Biol. Chem. 265: 15823-15831. Olivier L, Cadoret A, Duluc I, Capeau J, Gespach C, Cherqui G, Freund JN. (1999) Downregulation of the colon tumour-suppressor homeobox gene Cdx-2 by oncogenic ras. Oncogene 18:87-92. Olivier, M., Bautista, S., Valles, H. & Theillet, C. (1998) Relaxed cell cycle arrests and propagation of unrepaired chromosomal damage in cancer cell lines with wild-type p53. Molecular Carcinogenesis 23: 1-12. Olsen JH, Hahnemann JM, Barresen-Dale AL, et al. (2001) Cancer in patients with ataxia-telangiectasia and in their relatives in the nordic countries. J Natl Cancer Inst 93 (2): 121-7. Osteen, R.T., Henderson, I.C., Costanza, M.E., Wood, W.C., Harris, LR. (1986) In: Caner manual (Cady, B,. ed.), seventh edition, pp. 151-157., American Cancer Socitey. Massachusetts division, incorporated, Boston, MA. 200 Pages, G., Lenormand, P., L’Allemain, G., Chambard, J. C., Meloche, S., Pouyssegur, J. (1993) Mitogen-activated protein kinases p42 MAPK and p44 MAPK are required for fibroblast proliferation. Proc. Natl. Acad. Sci. USA 90: 8319-8328. Papadopoulos, N., Nicolaides, N. C., Wei, Y. F., Ruben, S. M., Carter, K. C., Rosen, C. A., Haseltine, W. A., Fleischmann, R. D., Fraser, C. M., Adams, M. D. et al. (1994) Mutation of a mutL homolog in hereditary colon cancer. Science. 263: 1625-1629. Parkin DM, Muir CS, Whelan SL, Gao YT, Ferlay J, et al. (1992) Cancer Incidence in Five Continents. Pub]. No. 120. Lyon, France: IARC Sci. Parodi, P. W. (1997) Cow's milk fat components as potential anticarcinogenic agents. J. Nutr. 127: 1055-1060. Parsons, PG, Musk, P, Goss, PD, Leah, J. (1983) Effects of calcium depletion on human cells in vitro and the anomaloud behavior of the human melanoma cell line MM170. Cancer Res. 43:2081-2087. Perry R. R., Kang Y., Greaves B. S. (1995) Effects of tamoxifen on growth and apoptosis of estrogen- dependent and -independent human breast cancer cells. Ann. Surg. Oncol. 2: 238-245. 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. Piazza, G.A., Rahm, A.K., Finn, T.S., Fryer, B.H., Li, H., Stoumen, A.L., Pamukcu, R & Ahnen, DJ. (1997) Apoptosis primarily accounts for the growth-inhibitory properties of sulindac metabolites and involves a mechanism that is independent of cyclooxygenase inhibition, cell cycle arrest, and p53 induction. Cancer Res. 57: 2452-2459. Pittelkow, M. R., & Scott, R. E. (1986) New techniques for the in vitro culture of human skin keratinocytes and perspectives on their use for grafting of patients with extensive burns. Mayo Clinic Proc. 61: 771-777. Pollard, M., Luckert, P. H., & Pan, G. Y. (1984) Inhibition of intestinal tumorigenesis in methylazoxymethanol-treated rats by dietary restriction. Cancer Treat. Rep. 68: 405-408. Potten, CS (1997) Epithelial cell growth and differentiation II: Intestinal apoptosis. American J. Physiology. 273:G253-G257. Potter JD, Slattery ML, Bostick RM, Gapstur SM (1993) Colon cancer: a review of the epidemiology. Epidemiologic Reviews 15(2): 499-545. 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. 201 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, V.R. (1978) Phenotypic diversity in experimental hepatomas: the concept of partially blocked ontogeny. The 10th Walter Hubert Lecture. Br.J.Cancer 38: 1-23. Potter, V.R (1987) Blocked ontogeny. Science 237: 964. Prasad KN, Carvalho E, Kentroti S, Edwards-Prasad J, La Rosa F, Kumar S, Freed C, Vemadakis A. (1994) Production of terminally differentiated neuroblastoma cells in culture. Restorative Neurol Neurosc 7: 13—19. Prasad KN, Cole WC, Hovland P. (1998) Cancer prevention studies: Past, present and future. Nutrition 14:197—210. Prasad KN. (1991) Differentiation of neuroblastoma cells: A useful model for neurobiology and cancer. Biol Rev 66:431—45 1. Prasad, K.N, Hovland, A.R., Nahreini, P., Cole, W.C., Hovland, P., Kumar, 8., Prasad, KC. (2001) Differentiation genes: are they primary targets for human carcinogenesis? Exp. Biol. Med. 226: 805-813. Preston, G.A., Lyon, T.T., Yin, Y., Lang, J .E., Solomon, G., Annab, L., Srinivasan, D.G., Alcorta, D.A. & Barrett, J.C. (1996) Induction of apoptosis by c-Fos protein. Mo].Cell Biol. 16: 211-218. Qiao, L., Harrif, R., Sphicas, E., Shiff, SJ. & Rigas, B. (1998) Effect of aspirin on induction of apoptosis in HT-29 human colon adenocarcinoma cells. Biochemical Pharmacology. 55:53-64. Radin, N. S., (2000) Apoptotic death by ceramide: will the real killer please stand up?. Medical Hypotheses. 57: 96-100. Rajan JV, Wang M, Marquis ST, Chodosh LA (1996) BRCA2 is coordinately regulated with Brcal during proliferation and differentiation in mammary epithelial cells. Proceedings of the National Academy of Sciences 93: 13078-13083, 1996. 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. Reddy, B. S. (1993) Dietary fat, calories, and fiber in colon cancer. Preventive Medcince. 22: 738-749. 202 Reddy, B. S., Wang, C. X., & Maruyama, H. (1987) Effect of caloric intake on azoxymethane-induced colon tumor incidence in male F 344 rats. Cancer Res. 47: 1226- 1228. Redman, C., Xu, M.J., Peng, Y.M., Scott, J.A., Payne, C., Clark, L.C., Nelson, M.A. (1997) Involvement of polyamines in selenomethionine induced apoptosis and mitotic alterations in human tumor cells. Carcinogenesis 18: 1195-1202. Reed, J. C. (1995) Bel-2: prevention of apoptosis as a mechanism of drug resistance. Hematol. Oncol. Clin. North. Am. 9:451. Reszka, A. A., Bulinski, J. C., Krebs, E. G., Fischer, E. H. (1997) Mitogen-activated protein kinase/extracellular signal-regulated kinase 2 regulates cytoskeletal organization and chemotaxis via catalytic and microtubule-specific interactions. Mol. Biol. Cell. 8,1219—1232. Rhyu, M. S. (1995) Telomeres, telomerase, and immortality. J. Natl. Cancer Inst. 87: 884-894. Riley RT, Wang E, & Merrill AH Jr. (1994) Liquid chromatographic determination of sphinganine and sphingosine: Use of the fiee sphinganine-to-sphingosine ratio as a biomarker for consumption of fumonisins. JAOAC. 77(2): 533-540. Rodrigues, N.R., Rowman, A., Smith, M.E., Kerr, I.B., Bodmer, W.F., Gannon, J.V. & Lane, DP. (1990) p53 mutations in colorectal cancer. Proc. Natl. Acad. Sci. USA. 87: 7555-7559. Rogers, A. E. (1997) Diet and breast cancer: studies in laboratory animals. J Nutr. 127: 933S-93SS. Rose DP. (1997) Dietary fatty acids and prevention of hormone-responsive cancer. Proc. Soc. Exp. Biol. Med. 216:224—33. Rozen P, Fireman Z, Figer A, Legum C, Ron E, Lynch HT. (1987) Family history of colorectal cancer as a marker of potential malignancy within a screening program. Cancer 60(2): 248-254. Russo J, Wilgus G, Russo 1H. (1979) Susceptibility of the mammary gland to carcinogenesis. I. Differentiation of the mammary gland as determinant of tumor incidence and type of lesion. Am J Pathol. 96:721—36. Russo J., Gusterson B. A., Rogers, A, E., Russo I. H., Wellings S. R., and Zwieten M. JV. (1990) Biology of disease, Comparative study of hmnan and rat mammary tumorigenesis. Lab Invest. 62: 244- 278. 203 Russo, J., Russo, I.H. (1987) Biological and molecular bases of mammary carcinogenesis. Lab.Invest. 57: 112-137. Sarensen, H.T., Olsen, J.H., Mellemkjaer, L., Thulstrup, A.M., Flemming, H.S., McLaughlin, I.K., Baron, J .A. (2000) Cancer risk and mortality in users of calcium channel blockers. A cohort study. Cancer. 89:165-170. Sakakura, C., Sweeney, E. A., Shirahama, T., Hagiwara, A., Yamaguchi, T., Takahahshi, T., Hakomori, S. I., Igarashi, Y. (1998) Selectivity of sphingosine-induced apoptosis. Biochemical-and-Biophysical-Research-Communications. (1998) 246:827-830. Sakakura, C., Sweeney, E. A., Shirahama, T., Ruan, F., Solca, F., Kohno, M., Hakomori, S. I., Fischer, E. H., Igarashi, Y. (1997) Inhibition of MAP kinase by sphingosine and its methylated derivative, N,N-dimethylsphingosine: A correlation with induction of apoptosis in solid tumor cells. International J. of Oncology. 11:31-39. Saklatvala, J., Rawlinson, L., Waller, R. J., Sarsfield, S., Lee, J. C., Morton, L. F., Barnes, M. J., Famdale, R W. (1996) Role for p38 mitogen-activated protein kinase in platelet aggregation caused by collagen or a thromboxane analogue. J. Biol. Chem. 271,6586-6589. Santana, P., Pefia, L.A., Haimovitz-Friedman, A., Martin, S., Green, D., McLaughlin, M., Cordon-Cardo, C., Schuchman, B.H., Fuks, Z., Kolesnick, R. (1996) Acid sphingomyelinase-deficient human lymphoblasts and mice are defective in radiation induced apoptosis. Cell. 86: 189-199. Satya Narayan, and Aruna S. J aiswal (1997) Activation of Adenomatous Polyposis Coli (APC) Gene Expression by the DNA-alkylating Agent N-Methle-nitro-N- nitrosoguanidine Requires p53. J. Biol. Chem. 272: 30619-30622. Savitsky K, Bar-Shire A, Gilad S, et al. (1995) A single ataxia telangiectasia gene with a product similar to PI-3 kinase. Science 268 (5218): 1749-53. Sawai, H., Okazaki, T., Yamamoto, H, Okano, H., Takeda, Y., Tashima, M., Sawada, H., Okuma, M., Ishikura, H., Umehara, H. & Domae, N. (1995) Requirement of AP-l for ceramide-induced apoptosis in human leukemia HL-60 cells. J. Biol. Chem. 270: 27326- 27331. Schalken, J. (1998) Molecular diagnostics and therapy of prostate cancer: new avenues. European Urology. 34:3S-6S. Schmelz E. M., Roberts P. C., Kustin E. M., Lemonnier L. A., Sullards M. C., Dillehay D. L., Merrill A. H. Jr. (2001) Modulation of Intracellular B-Catenin localization and intestinal tumorigenesis in vivo and in vitro by sphingolipids. Cancer Res. 61: 6723- 6729. 204 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 CFl 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., Kozutsurni, Y., Kawasaki, T. & Merrill, A. H., Jr. (1998) Induction of apoptosis by fumonisin B1 in HT -29 cells is mediated by the accumulation of endogenous fiee sphingoid bases. Toxicol. Appl. Pharm. 148: 252-260. Schmelz, E. M., Sullards, M.S., Dillehay, D.L. & Merrill, A. H., Jr. (2000) Colonic cell proliferation and aberrant crypt formation are inhibited by dairy glycosphingolipids in 1,2-dimethylhydrazine-treated CFl mice. J. Nutr. 130: 522-527. Schroy, P. C., Brown, S. S., Kim, K., Johnson, K. A., Murnane, M. J ., Yang, S., O'-Brien, M. J., Carney, W. P., Kupchik, H. Z. (1995) Detection of p21ras mutations in colorectal adenomas and carcinomas by enzyme-linked immunosorbent assay. Cancer 76: 201-209. Schubert, K. M., Scheid, M. P., & Duronio, V. (2000) Ceramide Inhibits Protein Kinase B/AKT by Promoting Dephosphorylation of Serine 473. J. Biol. Chem. 275: 13330- 13335. Seller, T. A. (1997) Genetic factors in the pathogenesis of breast cancer: their role and relative importance. J. Nutr. 127: 9293-932S. Senchenkov, A., Litvak, D. A., & Cabot, M. C. (2001) Targeting ceramide metabolism-a strategy for overcoming drug resistance. J. National Cancer Institute 93: 347-357. Separovic, D., He, J. & Oleinick, N.L. (1997) Ceramide generation in response to photodynamic treatment of L5178Y mouse lymphoma cells. Cancer Research. 57: 1717- 1721. 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. 205 Shay, J.W., Wright, W.E. & Werbin, H. (1993) Toward a molecular understanding of human breast cancer: a hypothesis. Breast Cancer Res.Treat. 25: 83-94. Shi, H. Y., Zhang, W., Liang, R., Kittrell, F., Templeton, N. S., Medina, D., & Zhang, M. (2003) Modeling human breast cancer metastasis in mice: maspin as a paradigm. I-Iistol. Histopathol. 18: 201-206. Shi, H. Y., Liang, R., Templeton, N. S., & Zhang, M. (2002) Inhibition of breast tumor progression by systemic delivery of the maspin gene in a syngeneic tumor model. Molecular Therapy. 5: 755-761. Slattery, M. L., Kerber, R. A.(l994) Family history of cancer and colon cancer risk: the Utah Population Database. Journal of the National Cancer Institute 86(21): 1618-1626. 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. Int]. J. Cancer. 73: 670- 677. Smith E. R., & Merrill AH Jr. (1995) Differential roles of de novo sphingolipid biosynthesis and turnover in the burst of fi'ee 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. Smith ER, Jones PL, Boss JM, Merrill AH Jr. (1997) Changing J774A.1 cells to new medium perturbs multiple signaling pathways, including the modulation of protein kinase C by endogenous sphingoid bases. J Biol Chem 272:5640-5646. Smith SA, Easton DF, Evans DG, Ponder BA (1992) Allele losses in the region 17q12-21 in familial breast and ovarian cancer involve the wild-type chromosome. Nature Genetics 2(2): 128-131, 1992. Sommerfeld, HJ, Meeker, AK, Piatyszek, MA, Bova, GS, Shay, JW, Coffey, DS (1996) Telomerase activity: a prevalent marker of malignant human prostate tissue. Cancer Research. 56:218-222. St John DJ, McDermott FT, Hopper JL, Debney EA, Johnson WR, Hughes ES. Cancer risk in relatives of patients with common colorectal cancer. Annals of Internal Medicine 118(10): 785-790, 1993. 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. II. Mechanisms. Cancer Causes and Control. 2: 427-442. 206 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. (19903) 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 (HL60) cells by serum factors and sphinganine. Cancer Res. 50: 222-226. Stubs, P. (1980) The anatomy of abuses. London, pp. 61. In: Royal College of Physicians: Medical Aspects of Dietary Fiber. Pitrnan Medical, Bath. Sun, M., Wang, G., Paciga, J. E., Feldman, R. I., Yuan, Z-Q., Ma, X-L., Shelley, S. A., Jove, R., Tsichlis, P. N., Nicosia, S. V., Cheng, J. Q. (2001) AKTl/PKB Kinase Is frequently elevated in human cancers and its constitutive activation is required for oncogenic transformation in NIHBT3 Cells. Am. J. Pathology. 159:431-437. Sun, W., Kang, K. S., Morita, I., Trosko, J. E., Chang, C-C. (1999) High susceptibility of a human breast epithelial cell type with stem cell characteristics to telomerase activation and immortalization. Cancer Res. 59: 6118-6123. Sweeney, E. A., Sakakura, C., Shirahama, T., Masamune, A., Ohta, H., Hakomori, S-I. & Yasuyuki, J . (1996) Sphingosine and its methylated derivative N, N—dimethlysphingosine (DMS) induce apoptosis in a variety of human cancer cell lines. Int. J. Cancer. 66: 358- 366. Swift M, Reitnauer PJ, Morrell D, et al. (1987) Breast and other cancers in families with ataxia-telangiectasia. N Engl J Med 316 (21): 1289-94. Tahara H, Nakanishi T, Kitamoto M, Nakashio R, Shay JW, Tahara E, Kajiyama G, Ide, T. (1995b) Telomerase activity in human liver tissues: comparison between chronic liver disease and hepatocellular carcinomas. Cancer Res. 56:218-222. Tahara, H., Kuniyasu, H., Yokozaki, H., Yasui, W., Shay, J. W., Ide, T., & Tahara, E. (1995a) Telomerase activity in preneoplastic and neoplastic gastric and colorectal lesions. Clinical Cancer Research. 1:1245-1251. Talapatra, S. & Thompson, C. B. (2001) Growth factor signaling in cell sm'vivalz Implications for cancer treatment. J. Pharmacology and Experimental Therapeutics. 298: 873-878. 207 Tang, 2., Shivapurkar, N., Frost, A., Alabaster, O. (1996) The effect of dietary fat on the promotion of mammary and colon cancer in a dual-organ rat carcinogenesis model. Nutr. Cancer. 25:151-159. Tannenbaum, A., & Silverstone, H. (1953) Nutrition in relation to cancer. Adv. Cancer Res. 1: 451-501. Telford. W.G., King, L.E., & Fraker, P.J. (1994) Rapid quantitation of apoptosis in pure and heterogeneous cell populations using flow cytometry. J. Immuno. Methods. 172: 1- 16. Tepper, C. L., Jayadev, S., Liu, H., 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 fiorn rat liver is capable of phosphorylating exogenous tyrosine-containing substances. Biochem. Intern. 18: 1203-1210. Testa, J. R. & Bellacosa, A. (2001) AKT plays a central role in tumorigenesis. Proc. Natl. Aca. Sci. USA. 98: 10983-10985. Tharnilselvan, V., Wei, L., Sumpio, B. E., & Basson, M. D. (2002) Sphingosine-l- phosphate stimulates human Caco-2 intestinal epithelial proliferation via p38 activation and activates ERK by an independent mechanism. In Vitro Cell. Dev. Biol. Animal. 38: 246-253. Thiagarajan, D. G., Bennink, M. R., Bourquin, L. D., Kavas, F. A. (1998) Prevention of precancerous colonic lesions in rats by soy flakes, soy flour, genistein, and calcium. Am. J. Clin. Nutr. 68:1394S-1399S. Thomson J A, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, Marshall VS, Jones JM. (1998) Embryonic stem cell lines derived fi'om human blastocysts. Science 282:1145—1147. Trock, B., 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. Trosko, J. E., Chang, C-C., Wilson, M. R., Upham, B., Hayashi, T., & Wade, M. (2000) Gap junctions and the regulation of cellular functions of stem cells during development and differentiation. Methods. 20:245-264. Trosko, J.E., Ruch, R.J. (1998) Cell-cell communication in carcinogenesis. Frontiers in Bioscience. 3 :d208-236. 208 Tseng, C.-J., Wang, Y.-J., Liang, Y.-C., Jeng, J .-H., Lee, W.-S, Lin, J.-K., Chen, C.-H., Liu, I.-C., & Ho, Y.-S. (2002) Microtubule damaging agents induce apoptosis in HL—60 cells and G2/M cell cycle arrest in HT -29 cells Tuyns, A. J ., Kaaks, R, Haelterman, M. (1988) Colorectal cancer and the consumption of foods: a case control study in Belgium. Nutr. Cancer. 11: 189-204. Uemura, H., Lin, Y. & Kubota, Y. (1998) Telomerase activity in association with the pathological differentiation of prostate cancer. Nippon.Rinsho. 56: 1287-1291. Uhrhammer N, Bay J O, Bignon YJ (1998) Seventh International Workshop on Ataxia- Telangiectasia. Cancer Res 58 (15): 3480-5. Van Noorden, C.J.F., Meade-Tollin, L.C., Bosman, ET. (1998) Metastasis: The spread of cancer cells to distant sites implies a complex series of cellular abnormalities caused, in part, by genetic aberrations. American Scientis. 86:130-141. Varmur, H. and Weinberg, RA. (1993) In: Gene and biology of cancer. W.H. Freeman Co. Chapter 2. pp38-39. Vaughn JP, Cirisano FD, Huper G, Berchuck A, Futreal PA, Marks JR, Iglehart JD (1996) Cell cycle control of BRCA2. Cancer Research 56(20): 4590-4594, 1996. Vaux, D.L. & Strasser, A. (1996) The molecular biology of apoptosis. Proc. Natl. Acad. Sci. USA. 93:2239—2244. Veer, P., Dekker, JM., Lanmars, JWJ., Kok, FJ., And Schouten, EG. (1989) Consumption of fermented milk products and breast cancer: a case-control study in The Netherlands. Cancer Res 49: 4020— 4023 Velie, E., Kulldorff, M., Schairer, C., Block, G., Albanes, D., Schatzkin, A. (2000) Dietary fat, fat subtypes, and breast cancer in postmenopausal women: a prospective cohort study. J. Natl. Cancer Inst. 92: 833-839. Verheij, M., Bose, R., Lin, X. H., Yao, B., Jarvis, W. D., Grant, S., Birrer, M. J., Szabo, E., Zon, L. I., Kyriakis, J. M., Haimovitz-Friedman, A., Fuks, Z., Kolesnick, R. N. (1996) Requirement for ceramide-initiated SAPK/JNK signalling in stress-induced apoptosis. Nature (London) 380,75-79. Vesper, H. Schmelz, E. M, Nikolova-Karakashian, M. N., Dillehay, D. L., Lynch, D. V., & Merrill, Jr. (1999) Sphingolipids in food and the emerging importance of sphingolipids to nutrition. J. Nutr. 129: 1239-1250. Vogelstein, B., Kinzler, K. W. (1992) p53 function and dysfunction. Cell. 70: 523. 209 Waddell, W.R. (1998) Stimulation of apoptosis by sulindac and piroxicam. Clinical Science (London) 95: 385-388. 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. Warden, L.-A., Menaldino, D.-S., Wilson, T., Liotta, D.-C., Smith, E.-R., & Merrill, A.- H. Identification of ammonium ion and 2,6-bis(omega-aminobutyl)— 3, 5- diiminopiperazine as endogenous factors that account for the "burst" of sphingosine upon changing the medium of J774 cells in culture. J Biol Chem 274:33875-33880, 1999. Wattenberg, L. W. ( 1977) Inhibition of carcinogenic efl‘ects 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. Weng, N. P., Levine, B. L., June, C. H., & Hodes, R. J. (1996) Regulated expression of telomerase activity in human T lymphocyte development and activation. J. Exp. Med. 183: 2471-2479. Westwick, J. K., Bielawska, A. E., Dbamboo, G. D., Hannun, Y. A., & Brenner, D. A. (1995) Ceramide activates the stress-activated protein kinase. J. Biol. Chem. 270:22689- 22692. Wiegandt, H. (1985) In: Glycolipids (W iegandt, H., ed.), pp. 199. Elsevier, New York. Wilkinson, M.G., Millar, J.B.A. (2000) Control of the eukaryotic cell cycle by MAP kinase signaling pathways. FASEB J. 14:2147-2157. Willet, W. C. (1997) Fat, energy and breast cancer. J. Nutr. 127: 921-923S. Willett W.C., Hunter D.J., Stampfer M.J., Colditz, G., Manson, J.E., Spiegelman, D., Rosner, B., Hennekens, CH. (1992) Dietary fat and fiber in relationship to risk for breast cancer: an 8-year follow-up. JAMA. 268:2037—2044. Willett, WC. (2001) Diet and Cancer: One View at the Start of the Millennium. Cancer Epidemiology Biomarkers and Prevention. 10: 3-8. William Kutchera, David A. Jones, Norisada Matsrmami, Joanna Groden, Thomas M. McIntyre, Guy A. Zimmerman, Raymond L. White, and Stephen M. Prescott (1996) Prostaglandin H synthase 2 is expressed abnormally in human colon cancer: Evidence for a transcriptional effect. PNAS.Vol. 93, Issue 10, 4816-4820. 210 Wolff, R. A., Dobrowsky, R. T., Bielawska, A., Obeid, L. M., & Hannun, Y. A. (1994) Role of ceramide-activated protein phosphatase in ceramide-mediated signal transduction. J. Biol. Chem. 269: 19605-19609. Woolf CM (1958) A genetic study of carcinoma of the large intestine. American J ournal of Human Genetics 10: 42-47. World Cancer Research Fund, American Institute of Cancer Research (1998) Food, Nutrition, and the prevention of cancer: a global perspective. Am. Inst. Cancer Res, Washington, DC. World Cancer Research Fund/American Institute for Cancer Research (1997) In: Food, Nutrition and the Prevention of Cancer: a global perspective. American Institute for Cancer Research. Wright WE, Piatyszek MA, Rainey WE, Byrd W, Shay JW. (1996) Telomerase activity in human gerrnline and embryonic tissues and cells. Dev Genet 18:173—179. Writing Group for the Women’s Health Initiative Investigators (2002) Risks and benefits of estrogen plus progestin in healthy postmenopausal women-principal results from the Women’s Health Initiative Randomized Controlled Trial. JAMA. 288: 321-333. Wu, A. H., Ziegler, R. G., Hom-Ross, P. L., Nomura-Abraham, M. Y., West, D. W., Kolonel, L. N., Rosenthal, J. F., Hoover, R. N., Pike, M. C. (1996) Tofu and risk of breast cancer in Asian-Americans. Cancer Epidemiol Biomarkers Prev 1996;5z901—906. Xia, z., Dickens, M., Raingeaud, J., Davis, R.J., Greenberg, ME. (1995) Opposing effects of ERK and JNK-p38 MAP kinase on apoptosis. Science. 270:1326-1331. Yang Q, Khoury MJ, Rodriguez C, Calle EE, Tatham LM, Flanders WD (1998a): Family history score as a predictor of breast cancer mortality: prospective data fiom the Cancer Prevention Study II, United States, 1982-1991. American Journal of Epidemiology 147(7): 652-659. Yang, D. D., Conze, D., Whitrnarsh, A. J., Barrett, T., Davis, R. J., Rincon, M., Flavell, R A. (1998b) Differentiation of CD4+ T cells to Th1 cells requires MAP kinase JNK2. Immunity 9,575-585. Yang, D. D., Kuan, C.-Y., Whitrnarsh, A. J., Rincon, M., Zheng, T. S., Davis, R. J., Rakic, P., F lavell, R A. (1997a) Absence of excitotoxicity-induced apoptosis in the hippocampus of mice lacking the Jnk3 gene. Nature (London) 389,865-870. Yang, H., Hsieh, C-Y., Sun, W., Chang, C.-C., & Schroeder, J.J. (Submitted) Chemopreventive and chemotherapeutic roles of sphingosine and ceramide in human breast cancer. 211 Yang, X., Khosravi-Far, R., Chang, H. Y., Baltimore, D. (1997b) Daxx, a novel Fas- binding protein that activates JNK and apoptosis. Cell 89,1067-1076. Yasumoto S, Kunimura C, Kikuchi K, Tahara H, Ohji H, Yamamoto H, Ide T, Utakoji T. (1996) Telomerase activity in normal human epithelial cells. Oncogene 13:433-439. Yoshimura, S.I., Banno, Y., Nakashima, S., Takenaka, K., Sakai, H., Nishirnura, Y., Sakai, N., Shimizu, S., Eguchi, Y., Tsujimoto, Y., Nozawa, Y. (1998) Ceramide Formation Leads to Caspase-3 Activation during Hypoxic PC 12 Cell Death: Inhibitory effects of Bel-2 on ceramide formation and caspase-3 activation. J. Biol. Chem. 273: 6921-6927. Yui J, Chiu CP, Lansdorp PM. (1998) Telomerase activity in candidate stem cells fi'om fetal liver and adult bone marrow. Blood 91 :3255—3262, 1998. Zanke, B. W., Boudreau, K., Rubie, E., Winnett, E., Tibbles, L. A., Zon, L., Kyriakis, J ., Liu, F .-F., Woodgett, J. R. (1996) The stress-activated protein kinase pathway mediates cell death following injury induced by cis-platinum, UV irradiation or heat. Curr. Biol. 6: 606-613. Zevenbergen, J. L., Verschuren, P. M., Zaalberg, J ., van Stratum, P., & Vles R. O. (1992) Effect of the amount of dietary fat on the development of mammary tumors in BALB/c- MTV mice. Cancer. 17: 9-18 Zhang H, Tombline G, Weber BL (1998) BRCAl, BRCA2, and DNA damage response: collision or collusion? Cell 92(4): 433-436, 1998. Zhang, L., Yu, J., Park, B.H., Kinzler, K.W., Vogelstein, B. (2000) Role of BAX in the apoptotic response to anticancer agents. Science. 290: 989-992. Zhang, W., Piatyszek, M.A., Kobayashi, T., Estey, E., Andreefl', M., Deisseroth, A.B., Wright, W.R. & Shay, J .W. (1996) Telomerase activity in human acute myelogenous leukemia: inhibition of telomerase activity by differentiation-inducing agents. Clin.Cancer Res. 2: 799-803. Zhu, X., Kumar, R., Manda], M., Sharma, N., Sharma, H.W., Dhingra, U., Sokoloski, J .A., Hsiao, R. & Narayanan, R. (1996) Cell cycle-dependent modulation of telomerase activity in tumor cells. Proc.Natl.Acad.Sci.U.S.A. 93: 6091-6095. Ziegler, R G., Hoover, R. N., Pike, M. C., Hildesheim, A., Nomura-Abraham, M. Y., West, D. W., Wu—Williams, A. H., Kolonel, L. N., Hom-Ross, P. L., Rosenthal, J. F., Hyer, M. B. (1993) Migration patterns and breast cancer risk in Asian-American women. J. Natl. Cancer Inst. 85: 1819—1827. 212 IIIIIIIIIIIIIIIIIIIIIIIIIIIIIII ' lllllllllllllull. lllll 293 0246