THE-as ”L MICHIGAN SYATEU l I! (NH! ll/llllllllll'ill/ll!!! 3 129 942739 ll ll This is to certify that the dissertation entitled Cloning and characterization of genes associated with the neoplastic transformation of human fibroblastic cells presented by Jing .Qing has been accepted towards fulfillment of the requirements for degree in Microbiology EM at Major professor Ph.D. nateAfilzr r117 0-12771 MS U is an Affirmative Action/Equal Opportunity Institution LIBRARY Michigan State University PLACE IN RETURN BOX to remove thie checkout from your record. To AVOID FINES Mom on or before dete due. DATE DUE DATE DUE DATE DUE MSU le An Affirmative Action/Equal Opponunlty lnetituion Willis-9.1 CLONING AND CHARACTERIZATION OF GENES ASSOCIATED WITH THE NEOPLASTIC TRANSFORMATION OF HUMAN FIBROBLASTIC CELLS By Jing Qing A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Department of Microbiology 1 997 ABSTRACT CLONING AND CHARACTERIZATION OF GENES ASSOCIATED WITH THE NEOPLASTIC TRANSFORMATION OF HUMAN FIBROBLASTIC CELLS By Jing Qing To understand the genetic changes involved in the malignant transformation of human fibroblast cells, I carried out differential mRNA display analysis, comparing the infinite life span, karyotypically stable, non-tumorigenic human fibroblast cell strain MSU-1.1 and one tumor-derived cell line, designated 6AISB1, which had been malignantly transformed by carcinogen treatment of MSU-1.1 cells. Five of the nine differentially expressed cDNA fragments identified by differential display were confirmed to be markedly downregulated in 6AISB1 cells by Northern blot analysis. DNA sequencing followed by a computer search of databases indicated that two of these were unique. One of the cDNAs corresponds to fibulin-1D, and the other one corresponds to a novel gene, designated 8T7. Northern and Western blotting analysis of 16 cell lines established from tumors formed in athymic mice by MSU-1.1-derived cell strains independently transformed in culture showed that 44% exhibited low level or lack of expression of fibulin-1D mRNA and protein. In a similar analysis of 15 malignant cell lines derived from patients, 80% showed low level or no expression of fibulin-1 D. To study the role of fibulin-1D in transformation, l transfected 6AISB1 cells and a human fibrosarcoma-derived cell line (SHAC) with a fibulin-1D cDNA expression construct. Transfectants displaying high levels of fibulin-1D were isolated and characterized. Elevated expression of fibulin-1D led to reduced ability to form colonies in soft agar and reduced invasive potential as tested in a matrigel in vitro invasion assay. Furthermore, expression of fibulin-1D resulted in a markedly extended latency in tumor formation in athymic mice. Using Northern analysis, I found that 10 out of 15 tumor cell lines derived from patients and 6 out of 16 cell lines established from tumors formed in athymic mice by MSU-1.1 cells transformed in culture have low or undetectable levels of $17 mRNA. Molecular cloning of a near full-length cDNA revealed that the novel gene encodes a putative transmembrane protein composed of 859 amino acids: the N-terminal domain consisting of 492 amino acids including a 5-fold cysteine-rich repeat of 40 amino acids homologous to the ligand binding repeat of the known low density lipoprotein receptor, a 24 hydrophobic amino acid stretch spanning the plasma membrane, and a C- terminal domain of 343 residues. The $17 gene is widely expressed in normal human tissues and is particularly abundant in human heart and skeletal muscle. Western blotting analysis using a specific anti-ST7 peptide antibody demonstrated that the levels of ST7 protein are high in normal fibroblasts and low in 12 sarcoma-derived cell lines tested. Altered expression of ST7 appears to be controlled at the transcriptional and posttranscriptional level. DEDICATION To my daughter, Julia Yaxin Wei for the magical, purest smile you give me everyday To my husband, Dong Wei without whom the research wouldn’t be so joyful To my parents, Shuhua Qing and Hanguo Li for their endless love and faith in me ACKNOWLEDGMENTS First and foremost, I would like to thank my major professor, Dr. J. Justin McCormick, for giving me the benifit of his wisdom, insight, encouragement, and never failing help during my years in the Carcinogenesis Laboratory. In addition, I wish I could use words to express my sincerest and deepest gratitude to Dr. Veronica M. Maher, for her advice, encouragement and lively discussions on science and other matters. Furthermore, I would like to give special thanks to the other members of my graduate guidance committee, Drs. Paul Coussens, Patrick Venta, and Richard Schwartz for their advice and invaluable time. Also I owe thanks to Terry McManus for his help in English proficiency in part of Chapter I. A million thanks go to the past and present members of the Carcinogenesis Liboratory for their generous friendship, assistance and stimulating discussions on science. They include Scott Boley, Clarissa Dallas, Philip Doroh, April Hoffman, Shixia Huang, Suzanne K. Kohler, W.Glenn McGregor, Lonnie Milam, Rebecca Odewaller, Sandra O’Reilly, Joseph Prezybewski, Beatrice Tung, Qingping Wang, Dong Wei, and Hong Zhang. TABLE OF CONTENTS LIST OF TABLES .................................................................................................. ix LIST OF FIGURES ................................................................................................ x INTRODUCTION ................................................................................................... 1 References ..................................................................................................... 5 CHAPTER I LITERATURE REVIEW A. Evidence that supports cancer is a genetic disease 1. Chromosomal abnormalities in cancer cells .......................................... 8 2. Familial cancers .................................................................................... 9 3. Defect in DNA repair and predisposition to cancer ................................ 10 4. Carcinogens and mutagens .................................................................. 11 B. The genetic elements governing cancer: Proto-oncogenes 1. Oncogene hypothesis ............................................................................ 12 2. Identification and isolation of oncogenes 2.1. Identification by homology with retroviral oncogenes ...................... 14 2.2. Identification by gene transfer ........................................................ 15 2.3. Identification by chromosome translocations .................................. 16 2.4. Identification by other routes 2.4.1. Identification by gene amplification ....................................... 18 2.4.2. Identification by structural homology with other oncogenes .................................................................. 19 3. Functions of proto—oncogenes 3.1. Growth factors as oncogenes ......................................................... 20 3.2. Protein tyrosine kinases as oncogenes 3.2.1. Receptor protein tyrosine kinases: EGF receptor as the prototype ................................................................... 22 3.2.2. Nonreceptor protein tyrosine kinases: Src as the prototype ................................................................... 23 3.3. Membrane associated G-proteins as oncogenes ............................ 24 3.4. SerlT hr kinases as oncogenes: Raf-1 as the prototype .................. 25 3.5. Nuclear transcription factors as oncogenes .................................... 26 3.6. Cell cycle kinase activators as oncogenes ..................................... 27 3.7. Inhibitors of apoptosis as oncogenes ............................................. 29 vi C. The genetic elements governing cancer: Tumor suppressor genes 1. Tumor suppressor gene hypothesis 1.1.The suppression of malignancy in cell hybrids ................................ 31 1.2. Familial cancer syndromes: Knudson's hypothesis ........................ 32 2. Identification and isolation of tumor suppressor genes 2.1. Identification by gene transfer ........................................................ 33 2.2. Identification by subtractive hybridization, differential hybridization and differential mRNA display ........................................................ 35 2.3. Identification by positional cloning 2.3.1. Localization of putative tumor suppressor genes 2.3.1.1. Localization by cytogenetics .................................... 37 2.3.1.2. Localization by molecular cytogenetics: Loss of heterozygosity ............................................ 38 2.3.1.3. Localization by molecular cytogenetics: Linkage analysis ..................................................... 38 2.3.1.4. Localization by molecular cytogenetics: Comparative genomic hybridization ......................... 39 2.3.2. Isolation of tumor suppressor genes .................................... 40 3. Function of tumor suppressor genes 3.1. Function of the p53 tumor suppressor gene ................................... 42 3.2. Function of the Rb tumor suppressor gene ..................................... 44 3.3. Function of the p16 tumor suppressor gene ................................... 45 D. Carcinogenesis is a multistep process: The role of oncogenes and tumor suppressor genes 1. Epidemiological studies of cancer .......................................................... 47 2. Experimental animal studies: Mouse skin carcinogenesis ...................... 47 3. Human cancer studies: Colorectal carcinogenesis ................................. 50 4. Transformation of human cells in culture 4.1. Immortalization of human cells in culture ........................................ 52 4.2. Neoplastic transformation of immortal human cells ........................ 55 LIST OF REFERENCES ................................................................................ 56 CHAPTER II SUPPRESSION OF ANCHORAGE-INDEPENDENT GROWTH AND MATRIGEL INVASION AND DELAYED TUMOR FORMATION BY ELEVATED EXPRESSION OF FIBULlN-1D IN HUMAN FlBROSARCOMA—DERIVED CELL LINES Abstract .......................................................................................................... 82 Introduction .................................................................................................... 83 Results Identification of fibulin-1 D as a differentially expressed mRNA ................. 85 Expression analysis of fibuIin-1D in multiple human tumor ceflfines ............................................................................................... 86 Establishment of SHAC and 6AISB1 clones stably expressing vii high levels of fibulin-1D protein ............................................................ 87 Anchorage-independence growth of the clones that express fibulin-1 D ......................................................................... 90 In vitro invasiveness offibulin-1Dtransfectants .......................................... 94 In vivo tumorigenicity of the fibulin-1 D expressing clones .......................... 94 In vitro proliferation offibulin-1Dtransfectants ........................................... 94 Discussion ...................................................................................................... 103 Materials and methods Cells and cell culture .................................................................................. 112 Growth rate ................................................................................................ 112 Differential mRNA display .......................................................................... 1 12 Northern blot analysis ................................................................................ 1 13 Preparation of cell lysates and conditioned medium .................................. 114 Western blot analysis of fibulin-1 ............................................................... 115 Plasmid construction and transfection ....................................................... 116 Anchorage independence assay ................................................................ 1 16 In vitro invasion assay ............................................................................... 116 Tumorigenicity assay ................................................................................. 1 16 Acknowledgment ............................................................................................ 1 18 References ..................................................................................................... 1 19 CHAPTER III CLONING AND CHARACTERIZATION OF A NOVEL GENE ENCODING A PUTATIVE TRANSMEMBRANE PROTEIN WITH ALTERED EXPRESSION IN HUMAN TUMORS Abstract .......................................................................................................... 123 Introduction .................................................................................................... 124 Results Identification of ST7 as a gene differentially expressed between preneoplastic and malignant human cells .............................. 126 Expression of ST7 in multiple human tumor cell lines ................................ 126 Molecular cloning of the full-length human ST7 cDNA ............................... 127 Primary structure suggests a novel transmembrane protein ...................... 130 Pattern of expression of ST7 mRNA in various human tissues ................. 136 Western blotting analysis of ST7 protein in human tumor cell lines .................................................................................... 136 Discussion ...................................................................................................... 146 Materials and methods Cells and cell culture .................................................................................. 149 Differential display ..................................................................................... 149 Northern blot analysis ................................................................................ 149 Cloning of human ST7 cDNA ..................................................................... 150 DNA sequencing and sequence analysis ................................................... 151 Production of anti-ST7 antibody ................................................................. 151 Western blotting analysis ........................................................................... 152 Acknowledgment ............................................................................................ 153 References ..................................................................................................... 154 viii LIST OF TABLES Chapter II Table 1. Human tumor-derived cell lines tested for fibulin-1 expression ................ 91 Table 2. Growth properties of parental cells, cells expressing a transfected-fibulin-1 D gene, and vector-transfected control cells ............ 102 LIST OF FIGURES Figure Chapter II 1. (A) Differential display comparing mRNAs from parental MSU-1.1 cell strain and its malignant derivatives cell line 6AISB1 ........................... 89 (B) Northern blot analysis confirming differential gene expression for fragment SG7 using the cloned cDNA fragment as a probe ................. 89 (C) Northern blot analysis using fibulin-1 D cDNA as a probe .......................... 89 (A) Northern blot analysis of fibulin-1 expression in multiple human tumor-derived cell lines .................................................................. 93 (B) RNA loading evaluated by a GAPDH cDNA probe ..................................... 93 . Western blot analysis of the expression (A) and secretion (B) of fibulin-1 D protein by normal cells and 10 tumor-derived cell lines under non-reducing conditions .......................................................... 96 . Western blot analysis of the expression and secretion of fibulin-1 D protein by Fibulin-1 D transfectants .............................................. 99 Colony formation by fibulin-1Dtransfected cell strains in 0.33% agarose ................................................................................ 101 In vitro invasiveness of fibulin-1 D transfected cell strains in matrigel assays ................................................................................ 105 . Tumorigenicity offibulin-1Dtransfected cell strains in athymic mice .................................................................................... 108 Chapter III (A) Differential display comparing mRNAs from parental MSU-1.1 cell strain and its malignant derivatives cell line 6AISB1 ........................... 129 (B) Northern blot analysis confirming differential gene expression for fragment ST7 using the amplified DNA fragment as a probe ................ 129 2. Northern blot analysis of ST7 expression in multiple human tumor-derived cell lines ................................................................ 132 x 3. Characterization of $17 cDNA clones (A) A diagram showing the four overlapping ST7 cDNA clones ............................................................................................ 134 (B) A near full-length cDNA sequence of ST7 gene and the deduced amino acid sequence ........................................................... 135 4. Alignment of the cysteine-rich repeat region of ST7 with that of the human LDL receptor ............................................................... 138 5. Expression of ST7 gene in normal human tissues ........................................... 141 6. Analysis of the specificity of the antibody B250 ............................................... 143 7. Western blot analysis of the expression of ST7 protein in normal human fibroblast cell lines and 12 tumor-derived cell lines .............................. 145 xi INTRODUCTION It is generally recognized that cancer development in humans is a multistep process, in which a series of genetic and epigenetic events lead to the emergence of a clone of cells that have escaped normal growth control (Peto, 1977; Father, 1984; Klein, 1987; Weinberg, 1989; Bishop, 1991; Hunter, 1991). Altered expression of proto-oncogenes and tumor suppressor genes as a result of mutations or other changes plays a pivotal role in the development and progression of tumors. For example, at least four or five genetic changes, including loss of function of tumor suppressor genes and activation of proto-oncogenes, are necessary for the development of colon carcinomas (Fearon and Vogelstein, 1990; Cunningham and Dunlop, 1996). In human breast cancers, mutations in a number of oncogenes, tumor suppressor genes and many other genetic loci are required before a cell becomes malignant (Jones et al., 1995). Despite the vast increase in our knowledge of oncogenes and tumor suppressor genes associated with human neoplasia over the past 15 years, the molecular events leading to the formation of most types of human tumors are still not well understood. A variety of genetic lesions are found in tumors, including point mutation, deletion, DNA amplification and chromosome rearrangement (Salomon et al., 1991; Lasko et al., 1991). Genes involved in cancer affect the normal functions of many cellular processes, including cell proliferation, senescence, apoptosis, cell-cell and cell-matrix interactions, DNA repair, invasion and motility, angiogenesis, and others (Sager, 1997). Yet very few cancer- related genes affecting these processes have been identified in human cancers. 1 2 Therefore, to understand the multistep nature of tumor formation, it is necessary to identify the genes that are involved in the transition of non-tumorigenic cells to malignant cells and establish any correlation between the disruption their functions and the tumor phenotype. The recent introduction of several in vitro transformation systems utilizing human cells in culture (Stoner et al., 1991; Reznikoff et al., 1993; McCormick and Maher, 1994; Rhim et al., 1994; Park et al., 1995) has facilitated the investigation of the cellular and molecular mechanisms involved in the multistep carcinogenic process. One of the advantages of these model systems is that they provide a means of dissecting the carcinogenic process under well-defined conditions. In our laboratory, transfection of the v-myc oncogene into a human neonatal foreskin-derived fibroblast cell line LG1 led to the establishment of a near-diploid, karyotypically stable, infinite life span human fibroblast cell strain MSU-1.1 (Morgan et al., 1991). MSU-1.1 cells are phenotypically normal and do not form tumors in athymic mice. Transfection of MSU- 1.1 cells with an activated ras oncogene expressed at high levels (Hurlin et al., 1989; Wilson et al., 1990) or treatment of MSU-1.1 cells with chemical carcinogens such as (_+_)-7B,8a-dihydroxy-9a,10a-epoxy-7,8,9,10-tetrahydrobenzo[a]pyrene (BPDE) (Yang et al., 1992) or gamma irradiation (Reinhold et al., 1996), followed by selection of focus-fanning cells, results in cells capable of forming tumors in athymic mice. However, the cellular genes responsible for the neoplastic transformation induced by these carcinogens remain poorly understood. One way to identify critical genes associated with malignant transformation is to analyze the gene expression profiles in normal and cancer cells with the same genetic background. Conventionally this is achieved using subtractive hybridization (Wieland et al., 1990; Lee et al., 1991) or differential hybridization (Steeg et al., 1988). The 3 recently developed differential mRNA display method (Liang and Pardee, 1992; Liang et al., 1993) involves the reverse transcription of the mRNAs followed by PCR amplification. The amplified cDNA fragments corresponding to the 3’ termini of mRNAs are separated on a DNA sequencing gel. The advantage of this method is that it allows one to display rapidly and simultaneously the expression of mRNAs from various phenotypically distinct variants. Therefore, genes that are overexpressed or downregulated can be identified at the same time. The objectives of the present study were (1) to identify genes that are differentially expressed between MSU-1.1 cells and one fibrosarcoma-derived cell line, designated 6AISB1, which had been malignantly transformed by carcinogen treatment of MSU-1.1 cells; (2) to determine whether the altered expression of identified genes is relevant in human tumor development in vivo by surveying the expression of the identified genes in tumor-derived cell lines from patients; (3) to clone the full-length cDNAs that correspond to the differentially expressed genes; (4) to define the biological function of the cloned gene(s) in tumor development by introducing the gene(s) into a proper recipient cell line and testing transformation-related phenotype. By identification, cloning and characterization of differentially expressed genes involved in the malignant transformation of MSU-1.1 cells, we should gain insight into the nature of the genetic changes underlying the neoplastic transformation of human fibroblasts in culture. We hope this information will provide useful diagnosis markers. It may also provide new approaches to therapy. Chapter I of the thesis reviews the literature that supports the mutation theory of the origin of human cancer. The various methods utilized to identify and isolate cellular oncogenes and tumor suppressor genes and the biochemical functions of these genes are discussed. Also discussed in Chapter I is the evidence that supports the multistep 4 hypothesis of carcinogenesis. Chapter II consists of a manuscript that will be published in the journal Oncogene. It describes the research I carried out which shows that fibulin-1 D, an extracellular matrix protein, is downregulated in 6AISB1 cells and a large fraction of fibrosarcoma—derived cell lines. Elevated expression of fibulin-1D in 6AISB1 cells and a human fibrosarcoma-derived cell line, SHAC, led to reduced ability to form colonies in soft agar and reduced invasive potential as tested in a matrigel in vitro invasion assay. Furthermore, expression of fibulin-1D resulted in a markedly extended latency in tumor formation in athymic mice. Chapter III consists of a manuscript that has been submitted to the journal Oncogene. It describes my research to identify and clone a novel gene, designated $17, which is downregulated in 6AISB1 and many other fibrosarcoma-derived cell lines. The deduced amino acid sequence suggests that ST7 encodes a putative transmembrane protein. Using a specific anti-ST7 peptide antibody, l demonstrated that the protein level of ST7 is high in normal human fibroblasts and low in sarcoma-derived cell lines. LIST OF REFERENCES LIST OF REFERENCES Bishop, J.M. (1991). Molecular themes in oncogenesis. Cell, 64:235-248. Cunningham, C., and Dunlop, MG. (1996). Molecular genetic basis of colorectal cancer susceptibility. Br. J. Surg., 83:321-329. Farber, E. (1984). Cellular biochemistry of the stepwise development of cancer with chemicals: G. H. A. Clowes memorial lecture. Cancer Res., 44: 4217- 4223. Fearon, ER, and Vogelstein, B. (1990). A genetic model for colorectal tumorigenesis. Cell, 61:759-767. Hunter, T. (1991). Cooperations between oncogenes. Cell, 64:249-270. Hurlin, P.J., Maher, V.M., and McCormick, J.J. (1989). Malignant transformation of human fibroblasts caused by expression of a transfected T24 H-RAS oncogene. Proc. Natl. Acad. Sci. USA, 86: 187-191. Jones, K. A., Brown, M.A., Solomon, E. (1995). Molecular genetics of sporadic and familial breast cancer. Cancer Surv., 25: 315-334. Klein, G. (1987). The approaching era of the tumor suppressor genes. Science, 238: 1539-45. Lasko, D., Cavenee, W., and Nordenskjold, M. (1991). Loss of constitutional heterozygosity in human cancer. Ann. Rev. Genet, 25: 281-314. Lee, S.W., Tomasetto, C., and Sager, R. (1991). Positive selection of candidate tumor suppressor genes by subtractive hybridization. Proc. Natl. Acad. Sci. USA., 88:2825- 2829. Liang, P., and Pardee, A. B. (1992). Differential display of eukaryotic messenger RNA by means of the polymerase chain reaction. Science, 257: 967-969. Liang, P., Averboukh, L., and Pardee, A. B. (1993). Distribution and cloning of eukaryotic mRNA by means of differential display: refinements and optimization. Nucleic Acids Res., 21:3269-3275. McCormick, J.J., and Maher. V.M. (1994). Analysis of the multistep process of carcinogenesis using human fibroblasts. Risk Anal., 14:257-263. 5 6 Morgan, T.L., Yang, 0., Fry, D.G., Hurlin, P.J., Kohler, S.K., Maher, V.M., and McCormick, J.J. (1991). Characteristics of an infinite life span diploid human fibroblast cell strain and a near-diploid strain arising from a clone of cells expressing a transfected v-myc oncogene. Exp. Cell Res., 197: 125-136. Park, N.H., Gujuluva, C.N., Baek, J.H., Cherrick, H.M., Shin, K.H., and Min, BM. (1995). Combined oral carcinogenicity of HPV-16 and benzo(a)pyrene: an in vitro multistep carcinogenesis model. Oncogene, 10: 2145-2153. Peto, R. (1977). Epideology, multistage models, and short term mutagenesis tests. In Origins of Human Cancer. Cold Spring Harbor Laboratory, Cold Spring Harbor, NY,1403-1428. Reinhold, D.S., Walicka, M., Elkassaby, M., Milam, L.D., Kohler, S.K., Dunstan, R.W. and McCormick, J.J. (1996). Malignant transformation of human fibroblasts by ionizing radiation. Int. J. Radiat. Biol., 69: 707-715. Reznikoff, C.A., Kav, C., Messing, E.M., Newton, M., and Swaminathan, S. (1993). A molecular genetic model of human bladder carcinogenesis. Semin. Cancer Biol., 4: 143-152. Rhim, J.S., Webber, M.M., Belle, 0., Lee, M.S., Amstein, P., Chen, LS, and Jay, G. (1994). Stepwise immortalization and transformation of adult human prostate epithelial cells by a combination of HPV-18 and v-Ki-ras. Proc. Natl. Acad. Sci. USA., 92: 11874- 11878. Sager, R. (1997). Expression genetics in cancer: Shifting the focus from DNA to RNA. Proc. Natl. Acad. Sci. USA, 94: 952-955. Salomon, E., Bozzow, J., and Goddard, AD. (1991). Chromosome aberrations and cancer. Science, 254: 1 153-1160. Steeg, P.S., Bevilacqua, G., Kopper, L., Thorgeirsson, U.P., Talmadge, J.E., Liotta, LA, and Sobel, ME. (1988). Evidence for a novel gene associated with low tumor metastatic potential. J. Natl. Cancer Inst, 80:200-204. Stoner, G.D., Kaighn, M.E., Reddel, R.R., Resan, J.H., Bowman, 0., Naio, 2., Matsukura, M., You, M., Galati, A.J. and Harris, CC. (1991). Establishment and characterization of SV40 T—antigen immortalized human esophageal epithelial cells. Cancer Res., 51: 365-371. Weinberg, RA (1989). Oncogenes, antioncogenes, and the molecular bases of multistep carcinogenesis. Cancer Res., 49: 3713-3721. VWeland, l., Bolger, G., Asouline, G., and nger, M. (1990). A method for difference cloning: gene amplification following subtractive hybridization. Proc. Natl. Acad. Sci. USA, 87: 2720-2724. Wilson, D.M., Yang, D.J., Dillberger, J.E., Dietrich, S.E., Maher,V.M. and McCormick, 7 J.J. (1990). Malignant transformation of human fibroblasts by a transfected N-ras oncogene. Cancer Res., 50: 5587-5593. Yang, D, Louden, C, Reinhold, D.S., Kohler, S.K., Maher, V.M. and McCormick, J. J. (1992). Malignant transformation of human fibroblast cell strain MSU-1.1 by (1)-78,8a- dihydroxy-9a,10a-epoxy-7,8,9,10-tetrahydrobenzo[a]pyrene. Proc. Natl. Acad. Sci. USA, 89: 2237-2241. CHAPTER 1 LITERATURE REVIEW A. Evidence that supports cancer is a genetic disease It is generally recognized that cancer is fundamentally a genetic disease, arising from changes in the DNA. Several lines of evidence support this theme: the detection of damaged chromosomes in cancer cells, the recognition of hereditary predisposition to cancer, the connection between cancer susceptibility and impaired ability of cells to repair damaged DNA, the evidence that relates the mutagenic potential of substances to their carcinogenicity, and the persuasive power of identified alterations in cancer-related genes. Here I describe some of the evidence in favor of the mutation theory of the origin of human cancer. 1. Chromosomal abnormalities in cancer cells In 1914, Boveri first formulated the somatic mutation hypothesis on the origin of human cancer (translation published in 1929). He proposed that the origin of cancer cells was due to the “wrongly combined chromosome complex occurring in a somatic cell and that this caused abnormal cell proliferation. The unlimited tendency to rapid proliferation in malignant tumor cells [could result] from a permanent predominance of the chromosomes that promote division...Another possibility is the presence of definite chromosomes which inhibit division...Cells of tumors with unlimited growth would arise if those inhibiting chromosomes were eliminated.” (Boveri, 1929). He proposed further that this defect was passed on to all cellular descendants of the original cancer cell. 8 9 The first chromosomal abnormality definitively associated with human cancer is the Philadelphia chromosome found in the leukemia cells of more than 90% of patients with chronic myelocytic leukemia. The Philadelphia chromosome results from a reciprocal chromosomal translocation between chromosome 9 and 22. So far, more than 100 commonly occurring translocations have been observed in Ieukemias, lymphomas and solid tumors (Solomon et al., 1991; Rabbitts, 1994). The consistent association of specific translocations with particular disease types, particularly when present as the sole chromosome abnormality, has led to the realization that these rearrangements identify critical genetic loci in the oncogenic process. In addition to chromosomal translocation, other chromosomal abnormalities found in cancer cells include chromosomal inversions, insertions, deletions, amplification and aneuploidy (Ruddon, 1995). Some of these aberrations occur consistently in certain specific cancer types. For example, deletions of 11p13 are found in Wilms’ tumors (Riccardi et al., 1990), and chromosome 9 monosomy is found in bladder adenocarcinoma (Solomon et al., 1991). These lead to the hypothesis that a specific locus is involved in a key way in generating these malignancies. It has also been found that certain chromosomal aberrations are common to tumors of different cellular origin. For example, the 3p13-23 region is commonly affected by deletions in small cell carcinomas and adenocarcinomas of the lung, renal cell carcinomas and ovarian adenocarcinomas (Dal-Cin and Sandberg, 1989). Deletions of the 1p11-22 region are found in melanoma, breast adenocarcinomas, and malignant fibrous histiocytomas (Solomon et al., 1991). These data suggest that a common locus must be inactivated for these tumors to develop. 2. Famllial cancers Inherited cancers represent a small fraction, perhaps 1% to 3% of total human 10 cancers (Knudsen, 1977). About 50 forms of hereditary cancers have been reported (Li, 1988; Birch, 1994). For example, dominant genetic inheritance accounts for about 40% of retinoblastoma and 20% to 40% of Wilms’ tumor. Familial adenomatous polyposis (FAP) of the colon is another example of a cancer transmitted as a Mendelian-dominant trait, with about 80% penetrance. Colorectal cancer will eventually develop in nearly 100% of untreated patients with FAP (Fearon and Johns, 1992). The Li-Fraumeni syndrome represents another hereditary neoplasia. Members of these affected families may develop a variety of cancers, including sarcomas, breast cancers, brain cancers, and leukemia at an early age. The inheritance pattern is that of an autosomal dominant trait with high penetrance (Garber et al., 1991). Collectively, these diseases suggest that a number of inherited predisposing mutations contribute to the causation of these cancer. The defective genes responsible for some of these diseases have been identified and cloned. For example, the Rb gene is responsible for retinoblastomas (Friend et al., 1986; Lee et al., 1987), the p53 gene for Li-Fraumeni syndrome (Srivastava et al., 1990), and the APC gene for FAP (Fearon and Johns, 1992). 3. Defect in DNA repair and predisposition to cancer One of the most convincing piece of evidence for a causal relationship between genetic mutations and cancer in humans comes from the fact that the incidence of cancer in patients with DNA repair deficiencies is greatly increased (Lindahl, 1994). In patients with certain recessively inherited disorders, such as xeroderrna pigmentosum (XP), ataxis telangiectasia, Fanconi’s anemia, Bloom’s syndrome, and hereditary nonpolyposis colorectal cancer (HNPCC), the frequency of specific tumors is significantly higher than in the general populations. Each of these disorders is characterized by the inability to repair specific kinds of physical or chemical damage to DNA. 11 XP is the most widely studied of the repair-deficient human diseases. Seven complementation groups of XP and one group that does not complement (XP variant) have been identified so far (Cleaver, 1990). These patients are characterized by extreme sensitivity of the skin to sunlight and usually suffer an age-specific incidence of skin cancer that is several thousand times higher than normal individuals. They typically develop multiple skin tumors that lead to death from metastatic squamous or basal cell carcinoma or malignant melanoma (Cleaver and Kraemer, 1989). All XP patients except XP variants are defective in nucleotide excision repair. Due to this defect, the cells of XP patients are less efficient in removing DNA lesions induced by DNA-damaging agents, for example, lesions caused by the ultraviolet light from the sun, and therefore are more prone to acquire genetic mutations than normal cells. Another well studied disease is HNPCC. Recently it has been found that the majority of HNPCC cases can be attributed to a defect in any one of the multiple loci responsible for DNA mismatch repair, for example, the hMSH2 (Fishel et al., 1993), hMLH1 (Papadcpoulos et al., 1994; Bronner et al., 1994), hPMS1 and hPMSZ (Nicolaides et al., 1994) genes. It has been proposed that the initial event in the development of tumors in HNPCC patients is the functional loss of a critical mismatch repair activity (Modrich, 1994), resulting in expansion or contraction of short nucleotide repeat sequences, which ultimately lead to mutations in cancer-related genes. For instance, the type II TGFB receptor gene, which is involved in negatively regulating epithelial cell growth, is mutated in many HNPCC tumors with the mismatch repair deficiency (Markowitz et al., 1995). The mutations found were either insertion or deletions in simple repeated sequences, which is consistent with defective mismatch repair. 4. Carcinogens and mutagens The evidence that chemicals can induce cancer in humans has been accumulating for 12 more than two centuries (Miller, 1978). Since most chemical carcinogens or their activated metabolites can react with DNA and cause mutations (Maher et al., 1968; Miller and Miller, 1981; Conney, 1982), the most plausible theory of carcinogenesis is that cancer is caused by genetic mutations. The mutagenicity of an agent can be assayed by various methods. For example, the Ames test (Ames et al., 1973; Maron and Ames, 1983) uses several strains of bacteria, Salmonella typhimurium, which are histidine auxotrophs and have poor nucleotide excision repair mechanism and an increased permeability to exogenously added chemicals. Using this system, Ames and his colleagues have estimated that about 90% of all carcinogens tested are also mutagens. Moreover, few non-carcinogenic agents show significant mutagenicity in this test system (McCann et al., 1975). Consistent with this mutation theory of carcinogenesis, chemical agents that cause genetic changes are frequently carcinogenic (Lawley, 1989). The extent of formation of some specific DNA adducts, for example, alkyl-OB-guanine, has been shown to quantitatively correlate with mutagenicity and carcinogenicity of nitrosamines and similar alkylating agents (Frei et al., 1978; Swenson et al., 1986). Polycyclic hydrocarbons, such as benzo(a)pyrene and 3- methylcholanthrene cause mutations in cultured Chinese hamster V79 cells, as measured by induction of resistance to 8-azaguanine and ouabain, and the degree of mutagenicity was related to the degree of carcinogenicity of the chemicals in vivo (Huberrnan and Sach, 1974; 1976). B. The genetic elements governing cancers: Oncogenes 1. Oncogene hypothesis Much of our understanding of the molecular mechanisms involved in cellular transformation comes from studies of tumor viruses. In 1910, Payton Rous first isolated a transforming agent from a spontaneous sarcoma formed in a Plymouth Rock chicken. 13 He demonstrated that tumor extracts still had the transforming ability after being passed through filters designed to exclude bacteria (Rous, 1911). Very little progress occurred on the identification of the agent until the late 1950s, when electron microscopic techniques enabled investigators to identify the infectious agent as a virus (Bernhard, 1960). In the 1960s and 1970s, the development of techniques of molecular virology led to the isolation and cloning of many animal tumor viruses, including those from mouse, chicken, cat and monkey (T ooze, 1980 and Weiss et al., 1982). Molecular characterization of these viruses revealed that they could be classified into two groups: one group having a genome composed of RNA and the other group having a genome composed of DNA (T ooze, 1980; Weiss et al., 1982). The RNA tumor viruses, named retroviruses, have a small RNA genome. Genetic analysis showed that, for many of these retroviruses, a viral gene(s) conferred transformed phenotype, i.e., increased refractivity or rounding of cells, or both, to host cells in culture. For example, several groups isolated temperature sensitive mutants of Rous sarcoma virus (RSV) that could induce morphological transformation of chick embryo fibroblasts at 36°C but not at the non-permissive temperature (Martin,1970; Friis et al., 1971; Kawai and Hanafusa, 1971). Golde (1970) and Vogt and colleagues (1970) showed that irradiation of certain strains of avian sarcoma virus resulted in the formation of viruses that retained the ability to grow but were no longer able to transform. Transforming genes were also found in DNA tumor viruses such as simian virus 40 and polyoma virus (Abrahams et al., 1975; Treisman et al., 1981; Bishop, 1985). These genes capable of inducing transformation were termed oncogenes (Bishop and Varmus, 1983). It was hypothesized that fragments of endogenous retrovirus genomes lay scattered throughout the mammalian genome as residues of ancient gerrnline infections by retroviruses. These fragmented viral genes might be able to transform the cell, once 14 activated by specific stimuli like those known to turn on latent proviruses (Huberrnan and Todaro, 1969). Even though this hypothesis is wrong, it led to our current insights into the molecular mechanisms of cancer development. 2. Identification and isolation of oncogenes By differential hybridization between a transformation competent RSV strain and a transformation defective RSV strain, Bishop and Vannus and their colleagues purified complementary DNAs corresponding to the transforming gene of RSV (c-src) (Stehelin et al., 1976a). Using a src cDNA as a probe, they carried out hybridization experiments with the genomic DNA of transformed chicken cells as well as normal cells and found that sequences homologous to this cDNA were present in both types of cells (Stehelin et al., 1976b). Later it was found that in normal salmon, mouse, calf and human DNA, there exist DNA sequences homologous to src (Spector et al., 1978a). RNA sequences corresponding to src were found in the cellular RNA of normal and neoplastic chicken cells, indicating that the gene is not only present but is also transcribed in both normal and transformed cells (Spector et al., 1978b). Further work extended this finding to many other retroviral oncogenes (Varmus, 1989). Searching for the cellular homologue of viral transforming genes was a fruitful way to uncover cellular oncogenes. Using the retroviral oncogenes as probes, more than twenty cellular oncogenes have been identified and cloned. A comparison of the cDNA sequence of the retroviral genes and intron-exon structure of the vertebrate genes makes it clear that these oncogenes were normal components of the genome, and that the retrovirus had transduced them (Bishop and Varmus, 1982). Analysis of multiple viral oncogenes and their homologues in animal genomes by DNA sequencing revealed that the viral genes often were mutated or altered substantially (Varmus, 1984; Bishop, 15 1985; 1987). The genes responsible for transformation in both naturally occurring and virus-induced tumors are designated oncogene, and their normal, unaltered cellular forms are called proto-oncogenes (Varmus, 1989; Bishop, 1991). 2mm In 1972, Hill and Hillavo showed that the DNA of cells transformed by RSV-infection was able to induce transformation of chicken embryo fibroblasts in culture as a result of transfer of the RSV genome. This finding led to the hypothesis that the DNA from cells transformed by chemical carcinogens or from cancer cell lines might also be able to transform normal cells. Weinberg and colleagues were the first to demonstrate that DNA from chemically transformed mouse fibroblasts were able to induce morphological transformation (focus formation) of NIH3T3 cells, an established non-neoplastic mouse fibroblast cell line (Shih et al., 1979). In 1981, several research groups reported that DNA from certain human tumor cell lines can also transform NIH3T3 cells (Shih et al., 1981; Krontiris and Cooper, 1981; Perucho et al., 1981). Cloning strategies were designed to rescue the dominant-acting human transforming genes from the transformed NIH3T3 cells. One method took advantage of the different repetitive sequences found in human and mouse cells. Alu sequences are highly repeated sequences (10 X 105 copies per genome) interspersed in the human genome and are present on average about once every 5 kb. During DNA transfection, approximately 10-30 kb of genomic DNA from the human tumor cells is transferred and integrated into the genome of the mouse cells. The detection of Alu sequences in the transformed NIH3T3 cells indicates that a human sequence has been taken up by the cells and may be contributing to the transformed phenotype. To eliminate human DNA that has nothing to do with the transformed phenotype, genomic DNA from NIH3T3 transforrnants was again transfected into normal NIH3T3 cells. Multiple rounds of 16 transfection and isolation of transformed cells lead to the enrichment of human sequences required for transformation of NIH3T3 cells. After enrichment, a genomic library was constructed using the DNA from NIH3T3 transforrnants. The DNA insert of human origin was identified by hybridization with Alu sequences and tested for transforming ability. This strategy led to the cloning of the res oncogene from the EJ human bladder carcinoma cell line (Shih and Weinberg, 1982). At the same time, the res oncogene was similarly cloned from several EJ-related bladder carcinoma cell lines (Santos et al., 1982; Der et al., 1982; Goldfarb et al., 1982). Many modifications of the transfection focus assay have been developed. One approach was to co-transfect tumor DNA with an antibiotic resistance marker gene. The drug-resistant clones were then selected in vitro, pooled and injected into athymic mice where tumor formation was monitored. This technique has revealed at least one new oncogene, mas (Young et al., 1986). Another approach makes use of an expression cDNA library instead of the fragmented genomic DNA. The cDNA library is constructed with mRNA from tumor cells and introduced into NIH3T3 cells to select for focus. Several potential oncogenes have been identified with this method (Miki et al., 1991; Chan et al., 1993; 1994). Such cloning experiments carried out with a large number of human tumor cell lines have revealed a diverse assortment of oncogenes (Varmus, 1984; Bishop, 1987; Miki and Aaronson, 1995). This method, however, suffers from several limitations. Among the most serious is its reliance on a single mesenchymal-derived recipient cell line which may not be susceptible to transformation by oncogenes from other cell types or to oncogenes which confer a property already acquired by NIH-3T3 cells (Varmus, 1984; Hunter, 1991). 23lll'fil'll | I I' 17 Specific chromosome translocations are often associated with distinct types of neoplasms (Heim and Mitelman, 1989; Soloman et al., 1991; Rabbitts, 1994). This observation implies that genes affected by the translocations might be instrumental in the genesis of the associated tumors. Based on chromosome translocations, a number of oncogenes have been identified. A good example is the cloning of the bcI-2 oncogene from B cell lymphoma cells. A common chromosomal abnormality that occurs in at least 90% of human follicular lymphomas is the reciprocal t (14;13) (q32 ; q21) translocation (Yunis et al., 1982). Since it was known that the immunoglobulin heavy chain gene (lgH) resides in chromosome band 14q32, several groups independently carried out Southern blot analysis using the lgH gene as a probe to test whether any chromosome rearrangements occurred in the lgH locus. Once they confirmed that this locus was mediating the translocation, genomic libraries were constructed from the DNA of follicular lymphoma cells carrying the t (14;18) chromosomal translocation, and screened with an lgH specific probe to isolate DNA fragments containing the breakpoint (T sujimoto et al., 1984; Cleary and Sklar, 1985). Subsequent chromosome walking experiments with chromosome 18 DNA fragments flanking the breakpoint ultimately led to the isolation of the novel oncogene, bcI-2 (T sujimoto et al., 1985; Cleary et al., 1986). This translocation resulted in the juxtaposition of the bcI-2 gene with the lgH gene, placing the bcI-2 upstream of the enhancer of the immunoglobulin gene and resulting in overexpression of the bcl-2 gene product (Graninger et al., 1987). Indeed several of the known cellular oncogenes have been found adjacent to interchromosomal breaks. For example, the human c-myc cellular oncogene, normally located at chromosome 8q“, is juxtaposed with the heavy, x, or k immunoglobulin (lg) genes from chromosome segments 14q32, 2p", or 22q11 in the well characterized 18 Burkitt’s lymphoma translocations (Taub et al., 1982; Dalla-Favera et al., 1983; Leder et al., 1983). These interchromosomal recombinations place the c-myc gene next to an lg locus and result in enhanced expression of the c-myc gene (Erikson et al., 1983; Taub et al., 1984). Another example is the 9:22 Philadelphia translocation present in almost all cases of chronic myelogenous leukemia (deKlein and Hagemeijer, 1984). The translocated gene on chromosome 9 is the c-abl oncogene (deKlein et al., 1982). The translocation results in the deletion of the amino-terminal portion of the abl gene product and the replacement of it with the amino-terminal of the bcr (for breakpoint cluster region) gene (Heisterkamp et al., 1985). This fusion protein exhibits elevated tyrosine kinase activity (Konopka et al., 1984). MW Many human tumors have amplified DNA sequences. The presence of amplified DNA sequences is often, although not always, manifested by the presence of chromosomes containing homogeneously staining regions (HSRs), or paired acentric chromatin bodies termed double minutes (DMs). Analysis of tumor cells and cell lines that exhibit DMs or HSRs has revealed the amplification of oncogenes. One such example is the isolation of the mdm-2 oncogene. A spontaneously transformed derivative of a mouse 3T3 cell line (designated 3T3-DM) contains an average of 25-30 DMs per cell. To isolate cDNA clones that represent genes amplified and overexpressed in the 3T3-DM cells, Cahilly-Snyder et al. (1987) constructed a cDNA library using mRNA from these cells. By differential hybridization with RNA from mouse cells lacking DMs or HSRs, they isolated two cDNAs representing amplified genes that were associated with the DMs, designated mdm-1 and mdm-2 (for mouse double minute). Later they demonstrated that enhanced 19 expression of the mdm-2 gene could induce tumorigenicity in NIH3T3 and Rat 2 cells (Fakharzadeh et al., 1991). WWW! Several of the oncogenes identified as described earlier belong to multigene families. It is attractive to assume that genes homologous to known oncogenes may also be candidates for oncogenes. For example, using probes derived from the c-myc oncogene, three other related genes have been identified, i.e., N-myc, L-myc and R-myc (Depinho et al., 1987). Furthermore, it has been shown that all four myc genes have transforming activity (Schwab et al., 1985; Depinho et al., 1987). Another example is the cloning of ras-related genes. Ha-, Ki-, and N-ras genes belong to a closely conserved family. Using ras probes, at least five more distantly related genes have been identified. One of these, R-ras (Lowe et al., 1987), has been reported to be mutated in human ovarian carcinoma cell lines and the mutated gene, when introduced into NIH3T3 cells, induced a fully malignant phenotype (Saez et al., 1994). 3. Function of oncogene products To date, more than 100 oncogenes have been identified. Extensive studies over the past two decades have revealed that proto-oncogene products are involved in the regulation of normal cellular growth and differentiation. Based on their biochemical functions, the protein products encoded by cellular oncogenes can be classified into seven groups. I will review examples in each category. 3.1. Growth factors as oncogenes In 1983, two groups of investigators reported that the sequence of the v-sis transforming gene, the acutely transforming oncogene of simian sarcoma virus, was homologous to the gene encoding the B chain of the platelet-derived growth factor (PDGFB) (Doolitle et al., 1983; Waterfield et al., 1983). Since then, several oncogene 20 products have been found to be growth factors. For example, oncogenes hst (Delli et al., 1987) and int-2 (Moore et al., 1986) encode fibroblast growth factor-related proteins. In general, growth factor stimuli are transmitted into the cell via specific transmembrane receptors that modify key regulatory proteins in the cytoplasm. These in turn affect the decisions controlling cell proliferation and differentiation, including changes in gene expression and reactivity to other factors. The relationship between growth factors and malignant transformation was first suggested by Spam and Todaro (1980) who proposed the autocrine model. This model proposes that growth factors and their cognate receptors are co-expressed by malignant cell populations, and that ligand- receptor interaction results in the autonomous proliferation of tumor cells. Several lines of evidence support this hypothesis. Expression of PDGF and its receptors has been reported in a high fraction of sarcomas as well as glioblastomas (Heldin and Westermark, 1989; Maxwell et al., 1990). Similarly, at least 70% of small cell lung cancer tumors and tumor-derived cell lines co-express the genes for stem cell factor and its receptor, the c-kit proto-oncogene (Krystal et al., 1996). Human colorectal carcinomas have been demonstrated to express a variety of growth factors, such as epidermal growth factor (EGF), transforming growth factor a (TGFa) and their cognate receptors, forming multi-autocrine loops (Shirai et al., 1995). These data suggest that aberrant production of secreted growth factors can play decisive roles in tumorigenesis by increasing the proliferation rate and degree of cellular autonomy (Aaronson, 1991; Cross and Dexter, 1991). 3.2. Protein tyrosine kinases as oncogenes The association of tyrosine phosphorylation with malignant transformation was first suggested by the discovery that many of the viral oncoproteins catalyzed the phosphorylation of proteins on tyrosine residues, and the level of phosphotyrosine in 21 proteins is elevated as much as 10—20 fold in cells transformed by these oncogenes (Hunter, 1987). To date, more than 40 protein tyrosine kinases have been identified. They can be classified into two groups: (1) Receptor protein tyrosine kinases, which span the plasma membrane with large extracellular and cytoplasmic domains; (2) cytoplasmic non-receptor protein tyrosine kinases, many of which are attached to the plasma membranes. 1 will describe an example for each category. :-.----o-"'uion-.n—.-u- -.-o-._ roe-to: Because growth factor receptor tyrosine kinases have the ability to generate a mitogenic signal, these molecules possess a latent oncogenic potential which, when activated, results in unregulated cell growth. In fact, several oncogenes are homologues of receptor tyrosine kinase. For example, the v-erbB oncogene is derived from the chicken EGF receptor gene (Yarden and Ullrich, 1988). The v-fms oncogene is derived from the gene for the receptor for the colony-stimulating factor 1 (Sherr, 1990). The EGF receptor (EGFR) is a 170 kDa transmembrane tyrosine kinase that is expressed on a wide variety of cell types. Other members in the EGFR family include p185neu tyrosine kinase (also known as HER2 or c-erbB-2), erbB-3 and erbB-4 (Schlessinger and Ullrich, 1992; Plowman et al., 1993). All four receptors possess a glycosylated, cysteine-rich, extracellular ligand binding domain, a single hydrophobic transmembrane region, and a cytoplasmic domain that contains a tyrosine kinase catalytic domain (Ullrich and Schlessinger, 1990). The interaction of a ligand with the EGFR or the p185neu/HER2 results in receptor dimerization, autophosphorylation (Ullrich and Schlessinger, 1990), and enhanced tyrosine kinase activity toward other substrates which elicit a mitogenic response in EGF-sensitive cells. Certain signaling molecules become physically associated and/or phosphorylated by the activated EGFR kinase. Those identified include the 22 phosphatidylinositol (PD-specific phospholipase C-y (PLC-y) (Rotin et al., 1992; Vega et al., 1992), the Pl-3'- kinase (PI3K), the PI4 kinase and the P15 kinase (Cochet et al., 1991), the GTPase activating protein (GAP) (Margolis et al., 1990), and the adaptor protein Shc (Pelicci et al., 1992). PLC4y is involved in the generation of two important messengers, inositol triphosphate and diacyl glycerol (Berridge and Irvine, 1989; Kikkawa et al., 1989). The former causes release of stored intracellular calcium and the latter activates protein kinase C (PKC). These secondary messengers appear rapidly in cells following stimulation with growth factors such as EGF. PKC belongs to a multigene family that encodes at least ten distinct isoforms (Nishizuka, 1992). Overexpression or gene alteration of certain isoforms of PKC has been reported to increase cell proliferation in culture (Housey et al., 1988; Hsiao et al., 1989) and induce neoplastic transformation (Cacace et al., 1993). The actions of a number of tumor promoters are thought to be mediated by PKC (Berridge and Irvine, 1989; Kikkawa et al., 1989). The importance of the PI3 kinase in transformation has been underscored by the recent identification of a new avian sarcoma virus, ASV16. The oncogene carried by ASV 16 encodes a Gag-PI3K p110 catalytic subunit fusion protein, which has PI3K activity (Hunter, 1997; Chang et al.,1997). The adaptor protein Shc has been shown to be a potent transforming gene when overexpressed (Pelicci et al., 1992). Constitutive activation of receptor protein tyrosine kinases can be achieved in a number of ways. In the case of the v-erbB oncogene, deletion of the extracellular ligand binding domain eliminates the negative control that this structure normally exerts on the cytoplasmic domain. A single point mutation in the p185neu transmembrane domain results in constitutive receptor oligomerization and activation (Weiner et al., 1989). Receptor-derived oncogenes possess other structural lesions such as point mutations and deletions in the cytoplasmic region, and carboxyl-terminal truncations that appear to 23 enhance and modulate the transforming signal (Woolford et al., 1988; Khazaie et al., 1989). However, these mutations seem to play a minor role in human cancer, since the most common cellular lesion found in human cancers involves autocrine activation (see 831) in association with receptor overexpression. Members of the EGFR family have been shown to be amplified or overexpressed in human tumors of several types, including lung cancer, breast cancer, ovarian cancer (Veale et al., 1987; Slamon et al., 1989), head and neck cancers (Grandis and Tweardy, 1993), glioblastomas (Nishikawa et al., 1994) and bladder cancer (Lipponen and Eskelinen, 1994). keg-[eeee‘l .".-,.._- ., .‘eeee- As noted above, Src was the first transforming oncogene discovered. At least nine members of the Src gene family have been discovered (Bolden et al., 1992; Brown and Cooper, 1996). Several were initially discovered as the normal counterparts of viral oncogenes. These proteins share structural similarities. They all have an extreme N- terrninal myristoylation signal presumably required for their attachment to the plasma membranes, the Src homology (SH) 3 and SH2 regions, a kinase domain, and a C- terrninal regulatory tail. The normal cellular functions of Src are still not clear. It has been shown that Src kinase is activated in PDGF-, CSF1-, EGF-, and FGF-stimulated fibroblasts (Brown and Cooper, 1996), and associates with the PDGF and CSF-1 receptors through its SH2 domain (Alonso et al., 1995). Activated Src protein has been shown to phosphorylate proteins that are implicated in mitogenic signaling pathways such as Pl-3 kinase (Gutkind et al., 1990; Haefner et al., 1995), GAP protein (Brott et al., 1991), and the adaptor protein, Shc (McGlade et al., 1992; Egan et al., 1993). These findings provide a biochemical link between the Src-family kinases and pathway utilized by activated growth factor receptors and suggest a role for Src in growth control. Therefore, v-Src 24 and activated c-Src may transform cells by upregulating mitogenic signaling pathways normally stimulated by growth factors. Another class of Src kinase substrates that have been identified includes proteins involved in cytoskeletal organization (e.g., vinculin), cell-substrate adhesion (e.g., fibronectin receptor), and cell-cell adhesion (e.g., focal adhesion kinase) (Reviewed by Brown and Cooper, 1996). The functional significance of phosphorylation of these proteins has yet to be established. 3.3. Membrane associated G-proteins as oncogenes Two forms of membrane associated G-proteins have been implicated in the regulation of cellular proliferation (Boume et al., 1990): the heterotrimeric G proteins and the monomeric products of the ras and related genes, including Ha-, Ki-, and N-ras. Recently, two more ras-like genes, T021 and R-ras have become potential targets for mutational activation in human cancers (Saez et al., 1994; Hunter, 1997). The normal function of ras family proteins is to serve as an ubiquitous signal transducer for multiple growth factor receptor tyrosine kinases (Crews and Erikson, 1993; Egan and Weinberg, 1993; McCormick, 1993; Marshall, 1995; Seger and Krebs, 1995). When a ligand such as EGF or PDGF binds to its receptor, the receptor is activated, triggering an autophosphorylation event. This leads, through intermediates such as adaptor proteins Shc and Grb2 (for growth factor receptor binding) to the guanine nucleotide exchanging factor 80$ (for son of sevenless), resulting in recruitment of Sos to the plasma membrane where ras is bound. The interaction between the guanine nucleotide releasing activity of Sos and ras leads to the release of GDP and binding of GTP to ras (Feig, 1993). This activates ras, which in turn leads to activation of a kinase cascade including Raf, Mek (also known as mitogen-activated protein kinase kinase or MAPKK), and MAP kinase (MAPK). The substrates of MAPK include various 25 molecules associated with cell growth control, including transcription factors, enzymes, and regulators of protein synthesis (Marshall, 1995). This signal transduction pathway is a crucial pathway regulating growth and differentiation in many cell types. In fact, mutations in the ras family of oncogenes are the most frequently detected alterations in oncogenes in both animal tumor model systems and in human cancers (Bos, 1989). Oncogenic forms of ras genes differ fromtheir normal counterparts by having a single mutation in codon 12,13, 59 or 61, which reduces their intrinsic GTPase activity and their ability to interact with GAP, thus keeping p21ras in the GTP-bound, activated mode (Tong et al., 1989; Krengel et al., 1990). Besides this well characterized ras-Raf-MAPK pathway, it has recently been shown that ras activation of Raf-MAPK independent pathways is sufficient to cause tumorigenic transformation (Khosravi-Far et al., 1996). Among the targets for ras required for transformation is Rac, a member of the Rho family of small G proteins. Mutant Rac has modest transforming activity, and a dominant negative mutant Rac blocks ras induced transformation (Khosravi-Far et al., 1995; Qiu et al., 1995). 3.4. SerlT hr kinases as oncogenes: Raf-1 as the prototype Raf-1 was first identified as the cellular homologue of the viral oncogene v-raf. The Raf family of oncogenes encodes serine/threonine-specific protein kinases. It comprises three members: Raf-1, A-Raf and B-Raf (Storm et al., 1990; Rapp, 1991). The three genes are dispersed over different chromosomes and have been mapped to sites that are frequently altered in human tumors (Storm et al., 1990). All three genes can be converted into oncogenes by N-terminal fusion, deletion or site-specific mutations (Rapp, 1991; Storm and Rapp, 1993). Raf proteins are believed to be key components of the growth factor receptor-ras-MAPK mitogenic signal transduction pathway (See 26 3.3.3.). Raf proteins can also be activated by phorbol ester through protein kinase C (Rapp, 1991; Stephens et al., 1992) or by G-protein coupled receptors (Cook et al., 1993; Howe and Maeshall, 1993). A number of studies also suggest that Raf-1 can elicit its oncogenic effect independent of MAPK activation. In Rat-1 cells, expression of a transfoming, activated Raf-1 does not stimulate MAPK activity and yet is sufficient to induce transformation (Gallego et al., 1992; Samuels et al., 1993). The mechanisms underlying the transformation are not clear. Raf-1 may also positively regulate cell growth by promoting progression of the cell cycle. It has been shown that Raf-1 can phosphorylate and activate the cdc25 phosphatases (Galaktionov et al., 1995). 3.5. Nuclear transcription factors as oncogenes Modulation of gene expression is frequently an important ramification of intracellular signaling and plays a vital role in the control of cell proliferation and differentiation. A number of nuclear transcription factors and transcription regulators have been implicated in different types of cancer (Lewin, 1991; Forrest and Curran, 1992). One example is the activator protein 1 (AP-1). AP-1 serves as the nuclear target of many oncogenic signal transduction pathways (Angel and Karin, 1991; Treisman, 1994). This multigene group includes members from the Fos (c-fos, fosB, fra-1, and fra-2) and Jun (c-jun, jun B, and jun D) families. Members of the Fos family proteins form heterodimers with Jun proteins via their bZip (basic region plus Ieucine zipper) regions. The complex binds to specific DNA sequences and regulates the transcription of genes containing the cis-acting element in their promoters. Both Fos and Jun were initially identified as retroviral oncogenes (Curran et al., 1982; 27 Maki et al., 1987). In keeping with their role in the control of cell proliferation, expression and activity of these genes are transiently stimulated by mitogenic signals. Rapidly dividing cells show increased levels of AP-1 transcriptional activation (Vogt, 1994). Studies using loss-of-function approaches, i.e., the expression of antisense RNA or the microinjection of antibodies showed that AP-1 complexes are required for theinitiation of DNA synthesis during S phase (Holt et al., 1986; Carter et al., 1991; Kovary and Bravo, 1991; Smith and Prochownik, 1992). This function may be mediated by regulating the transcription of cyclin D1 (Albanese et al., 1995; Phuchareon and Tokuhisa, 1995). Studies using transgenic mice showed that stable expression of c-fos led to dysregulation of bone growth, eventually resulting in osteosarcomas and chondrosarcomas (Ruther et al., 1987; 1989; Wang et al., 1991). Transgenic mice expressing an oncogenic form of jun developed fibrosarcomas at sites of wound healing (Schuh et al., 1990). A recent study with c-fos-null mice (Saez et al., 1995) demonstrated that c-fos is necessary for progression from benign papillomas to malignant squamous cell carcinomas and spindle cell tumors. Furthermore, activation of AP-1 activity has been documented in several types of human cancer, including human squamous cell carcinoma of the lung (Volm et al., 1992; Wodrich and Volm, 1993) and breast cancer (Walker and Cowl, 1991). Collectively, these studies demonstrate that altered activity of AP-1 complex could affect mitogenic control and promote neoplastic transformation of cells in specific tissues. However, AP-1 is not connected exclusively to positive growth signals (Sassone-Corsi, 1992). In a number of systems increased AP-1 activity is correlated with cessation of cell growth and differentiation (Vogt, 1994). The role of AP-1 as a growth stimulator or a growth attenuator is determined by additional, largely unknown factors. 3.6. Cell cycle kinase activators as oncogenes 28 In eukaryotic cells, progression through the cell cycle is strictly controlled by a family of cyclin proteins, the cyclin-dependent kinases (cdks), and kinase inhibitor proteins (Hartwell and Kastan, 1994; Hunter and Pines, 1994; Nurse, 1994). Unique combinations of cyclins and cdks assemble during each phase of the cell cycle. Theassociation of cyclins and cdks allows the subsequent activation of the complex and drives cell proliferation forward by phosphorylating specific substrates. The control of mammalian cell proliferation by extracellular signals occurs largely during the G1 phase of the cell cycle (Hunter and Pines, 1994; Hall and Peters, 1996). During the G1 phase, cells respond to extracellular signals by either advancing toward another division or withdrawing from the cycle into a resting state (Go) (Pardee, 1989; Sherr, 1994). Unlike transit through the S, 62, and M phases, G1 progression normally relies on stimulation by mitogens and can be blocked by antiproliferative agents. The D type cyclins (D1, DZ, and 03) and cyclin E are the primary G1 phase cyclins in mammalian cells (Sherr, 1993). Expression of the D type cyclins is rapidly induced after exposure of cells to mitogens; this expression declines when mitogens are withdrawn or antiproliferative agents are added (Sherr, 1994). Thus one might expect that the deregulation of cyclin D synthesis would make cell cycle progression less dependent on growth factors and contribute to oncogenesis. Recent studies have shown that, as a group, mutations in or deregulated expression of cell cycle genes constitute the most common genetic changes in tumor cells (Sherr, 1996), especially in one or another of the genes involved in controlling progression through the G1 phase of the cell cycle. The best characterized one is cyclin D1. Cyclin D1 is encoded by the CCND1 gene on chromosome 11q13 (Xiong et al., 1992). It was originally cloned as an oncogene, termed PRAD1, which was found to be activated by inversion of chromosome 11 in the genome of parathyroid adenoma cells 29 (Motokura et al., 1991; Rosenberg et al., 1991a). This inversion places CCND1/PRAD1 gene under the control of the parathyroid hormone gene promoter, resulting in overexpression of cyclin D1. In centrocytic lymphoma and multiplemyeloma, overexpression of cyclin D1 is achieved by a reciprocal translocation of chromosome 11q13 and the lgH locus on 14q34, bringing cyclin D1 gene under the influence of the lgH enhancer (Seto et al., 1992; Rosenberg et al., 1991b). Amplification and overexpression of cyclin D1 have also been documented in a broad spectrum of common human cancers, including breast cancers, squamous cell carcinomas of the head and neck, bladder cancers, small cell lung cancers and esophageal carcinomas (Hall and Peters, 1996). Consistent with the oncogenic role of cyclin D1 are observations that transgenic mice engineered to overexpress this cyclin in their breast tissues are prone to mammary adenocarcinomas (Wang at al., 1994), and that co- expression of cyclin D1 and myc genes in the lymphoid tissues of transgenic mice leads to rapid development of lymphomas (Bodrug et al., 1994; Lovec et al., 1994). 3.7. Inhibitors of apoptosis as oncogenes In the past few years, accumulating evidence suggests that an imbalance of homeostasis between cell growth and death plays a role in cancer development. Cancer cells usually acquire the ability to escape from programmed cell death (apoptosis) in response to DNA damage or other suboptimal conditions that would induce a normal cell to die. Bcl-2 family proteins are key regulators of apoptosis and are the best candidates implicated in carcinogenesis. As discussed earlier (see B.2.3), the bcI-2 gene was first identified as an oncogene involved in the development of human follicular lymphoma (T sujimoto et al., 1984). The t(14;18) translocation brings the bcl-2 gene under the control of the lgH enhancer, resulting in constitutively high expression of bcl-2 protein (Graninger et al., 1987). 30 The frequent association of a translocation involving the bcl-2 locus with cancer suggests that the bcl-2 gene may be an oncogene. Introduction of bcl-2 into aninterleukin 3-(IL-3)-dependent cell line led, not to factor-independent growth, but to factor-independent survival, and the cells were not tumorigenic (Vaux et al., 1988). Reed et al. (1988) reported that 3T3 cell constitutively expressing bcl-2 proteins only rarely became transformed. These studies were interpreted as indications that bcl-2 can contribute to tumorigenesis by allowing cells to survive, but malignancy requires additional genetic changes. Consistent with this view, the generation of bcl-2 transgenic mice using the lgH enhancer demonstrated that in the B cell lineage, deregulated cell survival resulted in dramatic polyclonal expansion of mature B lymphocytes and the development of diffuse immunoblastic B cell lymphomas. A high proportion of these tumors had rearrangements of the c-myc gene as a second genetic alteration (Strasser et al., 1990; McDonnell and Korsmeyer, 1991; Katsumata et al., 1992). Targeted overexpression of the bcl-2 gene in thymic lymphocytes, mammary and intestinal epithelium, and myeloid populations has not produced tumors (Hockenbery, 1994). One model for the function of the bcI-2 gene in carcinogenesis proposes that the bcI-2 gene acts synergistically with specific oncogenes to inhibit cell death under adverse growth conditions. This concept follows the recognition that several oncogenes have dual effects on cell proliferation and cell death. For example, expression of the c-myc gene promotes cell proliferation when growth conditions are favorable. However, when cells enter a growth arrest phase due to absence of growth factors or the action of cytostatic drugs, expression of c-myc induces apoptosis (Packham and Cleveland, 1995). C-myc-induced apoptosis has been shown to be blocked by overexpression of bcl-2 (Bissonnette et al., 1992; Evan et al., 1992; Fanidi et al., 1992). Additional cellular 31 oncogenes that can predispose cells to undergo apoptosis have been found, including c- fos and c-rel (Smeyne et al., 1993; Abbadie et al., 1993). Deregulation of the bcl-2 gene may also function as a survival mechanism in cancer cells predisposed to cell death by environmental factors. BcI-2 acts as a broad anti- apoptotic factor and opposes cell death following treatment with ionizing radiation and cancer drugs as well as hormone manipulations (Sentman et al., 1991; Miyashita and Reed, 1993). Several non-lymphoid neoplasms, including lung, prostate, colon and breast cancers, express high levels of bcI-2 protein (McDonnell et al., 1992; Pezzella et al., 1993; Bronner et al., 1994). C. The genetic elements governing cancers: Tumor suppressor genes 1. Tumor suppressor gene hypothesis The existence of tumor suppressor genes that can suppress or inhibit the growth of tumor cells is supported by three lines of evidence: (1) studies on somatic cell hybrids between normal and tumor cells; (2) familial cancer syndromes; and (3) the loss of specific chromosomes in cancers. 1 will focus on studies on somatic cell hybrids and familial cancer syndromes. 1.1. The suppression of malignancy in cell hybrids Somatic cell hybrid experiments provided the first evidence that genetic changes underlying the neoplastic transformation might result from the loss of function of specific genes. In these experiments, tumorigenic cells were fused with nontumorigenic cells, and the resulted hybrid cells were nontumorigenic (Harris et al., 1969; Harris, 1988). However, hybrids passaged in culture for extended periods were sometimes tumorigenic. Careful studies demonstrated that the tumorigenic segregants had lost one or more chromosomes from the initial hybrids (Harris and Klein, 1969; Stanbridge et al., 1981; Evans et al., 1982), and that suppression of malignancy in the hybrid cells 32 required the retention of specific chromosomes derived from the normal cells (Stanbridge et al., 1981; Klinger, 1982; Harris, 1988). This phenomenon of suppression holds true for a wide variety of cell or tissue types (Sager, 1985; 1989; Levine, 1993). Taken together, these experiments suggest that recessive genetic changes can be responsible for the tumorigenic phenotype and that gene products of normal alleles from normal cells are able to suppress tumorigenicity. By analyzing chromosomal loss in the tumorigenic variants of suppressed hybrids and using a microcell transfer technique, in which only one single chromosome derived from the normal cells is transferred to tumorigenic cells, experiments have identified and mapped the specific chromosomal location of some of these suppressor genes (Marshall, 1991; Levine, 1993). 1.2. Familial cancer syndromes: Knudson’s hypothesis A small proportion of most types of human tumors occur in familial form (Knudson, 1977; Li, 1988). Several well-defined tumor syndromes are inherited as simple autosomal dominant traits with high penetrance (Birch, 1994). Retinoblastoma, a rare pediatric eye tumor, is the prototype of this group. From the epidemiological studies of retinoblastoma, Knudson (1971) noted that 40% of retinoblastoma cases occurred in young children with a mean age of 14 months, and these tumors were often bilateral (originating in both eyes) with the mean number of tumors being three. In some cases there was a family history of this disease. If these patients were cured through surgical intervention, they often had a substantially increased risk of developing other malignancies, especially osteosarcoma. These observations suggested an inherited component for these cancers. Howevr, about 60% of the retinoblastoma cases didn’t fit this pattern. In these cases, no familyhistory of cancers was noted. These tumors were typically first detected at a later age, and were 33 always unilateral (affecting only one eye) with one cancer per patient. This class of retinoblastomas was quite rare, occurring in about one in 30,000 people. Following the earlier suggestions by De Mars (1970), Knudson (1971) proposed a hypothesis to explain the origins of these two categories of retinoblastoma. He suggested that children who develop retinoblastomas at an early age inherited a mutant allele of a gene, later called the retinoblastoma susceptibility gene or Rb, and that a second somatic mutation in this gene in a retinoblast would then give rise to this cancer. This was a common event, which was consistent with an average of three independent cancers per patient and occurred in 90% of such patients. In contrast, children who develop retinoblastoma at a later age did not inherit a mutant Rb allele, and two independent mutations of this gene in a single retinoblast must have occurred to give rise to a tumor since the Rb gene is recessive at the cellular level. This was consistent with the low frequency (one in 30,000 people). Predisposition to retinoblastoma was autosomally dominant because at typical mutation rates almost everyone who has inherited one defective Rb gene will sustain a second mutation in the Rb gene in at least one retinoblast. This unifying hypothesis leads to the concept of genes whose product normally prevents cancerous growth and both alleles must be lost via mutations for it to play a causal role in cancer development. 2. Identification and isolation of tumor suppressor genes 2.1. Identification by gene transfer Unlike the dominant oncogenes, which can be identified with relative ease, assaying directly for tumor suppressor gene function is difficult. It is easy to identify a small number of transformed cells admist a background of normal cells because thetransformed cells have a growth advantage. In contrast, identifying a small number of normal (revertant) cells which grow like normal cells in a background of transformed 34 cells is not a trivial task. Based on observations that alteration in certain phenotypes in vitro is associated with loss of malignancy in vivo, several groups devised assays to clone tumor suppressor genes directly using a gene transfer technique (Noda, 1990). In the course of characterizing the in vitro phenotypes of spontaneously nontumorigenic revertants from the v-Ki-ras-transformed NIH3T3 cells, Noda and colleagues found that flat revertants exhibited greatly reduced malignancy in vivo (Noda et al., 1983; Basin and Noda, 1987). They used “flatness” as an in vitro marker to clone tumor suppressor genes. They transfected the tumorigenic DT cell line (a subline of v- Ki-ras-transformed NIH3T3 cell) with a normal human foreskin fibroblast expression library, isolated flat revertants, and recovered the transfected cDNA, Krev-1, from the genome of the revertant cells. When Krev-1 was introduced into DT cells and expressed at high levels, some of the transformed phenotypes, e.g., the efficiency of colony formation and the size of the colonies in soft agar, were reduced. The Krev-1 gene encodes a protein homologous to the p21ras protein (Kitayama et al., 1989). Since H-ras transformed rat FE-8 cells showed an increased sensitivity toward ouabain when compared to their normal counterparts (Noda et al., 1983), Schaefer et al. (1988) established a functional assay to identify and clone DNA sequence capable of suppressing neoplastic transformation. Genomic DNA from normal human placenta was introduced into FE-8 cells by co-transfection with a plasmid conferring drug resistance. Drug-resistant cells were subjected to treatment with ouabain. The surviving clones lost the morphology of transformed cells, acquired the ability to grow in an anchorage- dependent manner only, and showed reduced ability to form tumors in athymic mice.Using the human Alu repetitive sequences as probes, the suppressor gene in a secondary revertant clone was isolated. These cloning experiments are largely based on alterations in certain phenotypes in 35 vitro that correspond well to loss of malignancy in vivo. The problem is lack of an efficient selection procedure. Also, the spontaneous revertants will give rise to false positives. Because of these limitations, gene transfer is not an efficient method to clone tumor suppressor genes. 2.2. Identification by subtractive hybridization, differential hybridization and differential mRNA display Since malignant transformation is caused by accumulation of diverse genetic changes, including mutations and/or altered expression of critical genes (Sager, 1997), gene transfer experiments may not detect genes that are required but are not sufficient to suppress the malignant phenotype. Therefore, identification and cloning of genes differentially expressed between tumor cells and well matched normal counterparts, especially those genes that are downregulated in the tumor cells, may uncover new tumor suppressor genes. Currently, several methods are available for this purpose, namely, subtractive hybridization, differential colony hybridization and differential mRNA display. Subtractive hybridization is designed to select for genes expressed uniquely or preferentially in one of a pair of closely related cell populations (Sargent, 1987). cDNA synthesized from the mRNA of one cell population is hybridized with excess amount of mRNA from the other cell population. The cDNA-RNA hybrids representing mRNAs that are equally expressed in both cell populations are efficiently separated from the unpaired cDNAs by hydroxyapatite chromatography. The unpaired cDNAs from the first round of hybridization are hybridized again with excess amount of mRNA from the other cell population. Usually the unpaired cDNAs from the second round of hybridization represent mRNAs that are uniquely expressed in the cell of interest. These cDNAs are inserted to a vector to construct a subtractive library or are used as probes to screen a 36 library. Using such a strategy, Sager and colleagues isolated several putative tumor suppressor genes involved in the development of breast cancer (Lee et al., 1991 ). The technique of differential colony hybridization has been widely used to identify mRNAs regulated by cell differentiation, growth and cytokine treatment (Leonard et al., 1987; Sarrna et al., 1992; Femandez-Pol et al., 1993; Green et al., 1995). To identify genes uniquely expressed in metastatic cells, Steeg et al. (1988) utilized a series of related murine melanoma cell lines of varying metastatic potential. These cell lines are derived from K-1735 cell line. A cDNA library was constructed using mRNA from K- 1735 cells. Duplicate filters of the library were prepared and one was hybridized to labeled cDNA from a low-metastatic cell line and one to cDNA from a high-metastatic cell line. Using this approach, they isolated a novel gene, termed nm23, whose expression level was inversely correlated to tumor metastatic potential (Sobel, 1990). The major drawbacks of this techniques are poor sensitivity and difficulty in isolating the relatively few positive clones from the high background hybridization (Cochran et al., 1987). The differential mRNA display procedure developed by Liang and Pardee compares the profile of gene expression in closely related eukaryotic cells by systematically amplifying the 3' end of mRNAs using reverse transcription and PCR ( Liang and Pardee, 1992; Liang et al., 1993). It consists of two steps. First, a subset of the mRNA populations are reversely transcribed using an anchored oligo(dT) primer; secondly, the3' end of mRNAs are amplified by PCR using an arbitrary 10mer primer and an anchored oligo(dT) primer. The PCR products are separated on a DNA sequencing gel. By comparing the band patterns from two cell populations, mRNAs that are upregulated or downregulated can be identified simultaneously. Compared with the conventional subtractive or differential hybridization method, differential mRNA display provides a 37 rapid and efficient approach to detect differentially expressed mRNAs from pairs of phenotypically distinct cells of the same genetic background. Using this approach, Sager and colleagues have isolated more than 50 novel genes which are no longer expressed or are strongly downregulated in breast carcinoma cell lines (Sager, 1997). Genes that are downregulated in other types of cancer have also been identified with this method (Sun et al., 1994; Simon et al., 1996). 2.3. Identification by positional cloning A powerful way to identify and clone tumor suppressor genes is positional cloning, whereby a gene associated with certain types of cancer is isolated on the basis of its approximate chromosomal position (Collins, 1991; Wrcking and lMlliamson, 1991). The first step in this approach is the mapping of the gene to a particular human chromosome. Then various strategies can be used to identify and characterize the specific gene in that genomic region. I will describe the various methods available for localization and cloning of such genes. 2311 I' I' [III 23111 III | l' Cytogenetic studies in solid tumors have generally yielded a plethora of inconclusive data but there have been some consistent findings that have led to the isolation of new tumor suppressor genes. For example, about 3% to 5% of hereditary retinoblastomacases were found to be associated with gross abnormalities in chromosome 13 (Gallie et al., 1990). Other studies found patients who had small interstitial deletions of 13q (Franke, 1978; Yunis and Ransay, 1978). In all cases studied, the deletion involved band 13q14.1. This regional deletion was also demonstrated in some sporadic retinoblastomas (Balaban et al., 1982). These studies suggested that the putative Rb tumor suppressor gene resided within 13q14.1. 38 Cytogenetic studies also helped to localize the tumor suppressor gene of Wilms’ tumor on chromosome 11p13 (Riccard et al., 1990). However, most of the time, the regional chromosome loss is too small to be detected at the visible chromosome level. Molecular cytogenetics provides a more sensitive method to detect chromosome abnormality by analysis of extracted DNA. Loss of heterozygosity (LOH) involves comparing normal and tumor tissues from the same patient to detect allele imbalance at polymorphic loci. The typical situation is that the two alleles from the non-tumorigenic cells of the body contain a restriction fragment length polymorphism (RFLP), while the cells from the tumor tissue have lost one of these two alleles. By using panels of RFLPs that map to different regions of a given chromosome, and studying LOH in enough tumors, a common region of deletion can be identified. LOH could arise from a loss of a chromosome, a small deletion of the genetic locus, or a gene conversion by homologous recombination. Typically a mutation arises in a tumor suppressor gene and this is followed by LOH at that locus, thus eliminating both wild-type alleles. Besides RFLPs, microsatellite sequences are particularly informative for detection of LOH. They are extremely polymorphic so that most individuals are constitutionally heterozygous (Weber, 1990). Furthermore, they can be detected by PCR and are not dependent on the presence or absence of a restriction enzyme site. For those genes associated with hereditary predisposition to some form of cancer, genetic linkage analysis in affected families provides an additional route to localization. This technique depends on the crossover between homologous chromosomes at meiosis which means that DNA sequences, even on the same chromosome arm, will 39 not be transmitted together over several generations unless they are physically very close together. If homologous sequences on the two parental chromosomes are polymorphic, meiotic reassortment can be detected. The purpose of linkage analysis is to identify the close physical neighbors of the gene of interest in the genome (Yates and Connor, 1986). This involves finding a large number of families in which multiple individuals are affected with a specific cancer, and the testing of the individuals in these families with a large panel of DNA markers that show polymorphism. When a marker that tends to be co-inherited with the cancer gene in all (or almost all) affected members of those families is found, it indicates that the cancer gene is located near to that marker and thus allows mapping of the gene to that chromosome. Linkage analysis then proceeds by testing additional markers from the same chromosome arm and measuring the frequency of meiotic crossover between each of them and the cancer trait. In this way, the relative positions of a number of DNA sequences (including the putative cancer gene) can be established to facilitate gene isolation. The major limitation of linkage analysis in identifying tumor suppressor sequences is that it can only be used to study familial cancers and requires relatively large, well-defined pedigrees. Il" _—_ 1"‘I‘. 'll'z-‘Q‘l'll I 'lQ-‘l In comparative genomic hybridization (CGH), a single hybridization allows DNA copy number changes in the entire genome of a tumor to be assessed in comparison with normal tissue DNA (Kallioniemi et al., 1992). The principle of CGH is simple. Representative genomic DNA is prepared separately from tumor and normal tissue of the same patient. The DNAs are labeled with two different fluorochromes, e.g., one with a green fluorochrome, and the other with a red. Equal amounts of these labeled DNAs are mixed and simultaneously hybridized to normal human metaphase spreads. The relative amounts of tumor and normal DNA probes bound at a given chromosomal locus 40 depend on the relative abundance of these sequences in the two DNA samples and can be quantitated by measurement of the ratio of green to red fluorescence along the length of each chromosome. The normal DNA probes serve as a control for local variations in the ability to hybridize to the target chromosome. Therefore, where there has been either a deletion or an amplification of DNA in the tumor, the green-to-red ration will deviate from the norm. This method has led to the localization of several susceptibility loci for human cancers (Hemminki et al., 1997; Kallioniomi, 1997). MW Once the approximate location of a putative tumor suppressor gene is mapped within two flanking markers, several methods can be used to clone the gene. The first method is the candidate gene method. If there are one or several known genes in the region of interest, these then become “candidates” for the gene causing the cancer in question and are examined, for any mutations that segregate with the disease in affected families and for a putative function that may plausibly account for the observed phenotype. This procedure led to the identification of the p53 tumor suppressor gene. By using multiple probes on the short arm of chromosome 17, Baber et al. (1989) defined a region that was commonly lost in colon carcinomas. This region contains the site of the p53 gene. The p53 protein was implicated in tumorigenesis because of its interaction with the transforming large T antigen of simian virus 40. Since no alteration in the remaining p53 locus could be detected in tumors, the p53 cDNA from tumors that had lost one p53 allele was sequenced. This analysis with two tumor samples found the presence of single point mutations occurring in evolutionarily conserved regions (Finlay et al., 1988). Additional cDNA sequencing showed that many different human tumors commonly contained mutant p53 proteins (Hollstein et al., 1994; Lane, 1994). 41 The second method is chromosome walking. This technique was used by Dryja et al. (1986) to clone the Rb tumor suppressor gene. They showed that a chromosome 13 sequence that mapped to q14 detected a region that was homozygously deleted in the tumor DNA from two unrelated Rb patients. This sequence was then used in a chromosome walking to identify a DNA fragment conserved in mouse and human, which suggested that it contained a coding sequence. This DNA fragment identified a 4.7 kb mRNA, which was expressed in normal retinal cells, but was altered or not expressed in retinoblastomas (Friend et al., 1986; Lee et al., 1987) Another approach involves a direct search for transcribable sequences. Currently two methods are frequently used: hybridization selection and exon trapping (Duyk et al., 1990; Nehls et al., 1994; Datson et al., 1996). With hybridization selection, one first screens a genomic library with the marker sequences believed to lie close to the gene of interest. Then one screens a cDNA library from an appropriate tissue with the labeled genomic clone to isolate the cDNA. With exon trapping, genomic DNA of interest is partially digested with a restriction enzyme and cloned into a vector containing a splice donor and an acceptor sites positioned flanking the inserted genomic DNA. RNA transcripts derived from such vectors are processed in vivo and exons contained within the inserted genomic fragments become flanked by known sequences in the resulting mRNAs. Reverse transcription-coupled PCR can then be used for subsequent cloning and sequence analysis of trapped exons. Identification of cDNA by either method results in a candidate tumor suppressor gene. Confirmation that it is the tumor suppressor gene usually requires that mutations that segregate with cancer patients can be identified in that gene or that its biological function can account for the phenotype. 3. Function of tumor suppressor genes So far, about a dozen confirmed tumor suppressor genes have been cloned (Sager, 42 1997). Their biological functions range from transcriptional regulation to cell cycle check point control. I will describe the functions of three tumor suppressor genes that have been extensively studied, namely p53, Rb and p16. 3.1 . Function of the p53 tumor suppressor gene More than 50% of human tumors contain mutations in the p53 gene (Hollstein et al., 1994). The p53 tumor suppressor gene encodes a 393 amino acids nuclear phosphoprotein. The protein has three structural and functional domains. The N- terrninal region of p53 protein is highly charged and acts as a transcription activation domain. It interacts with the basal transcription machinery to regulate gene expression (Lu and Levine, 1995). p53 transcription activation is negatively regulated by MDM2 protein which can act as an oncoprotein (Lin et al., 1995). The core or central region of p53 is highly conserved in evolution and has sequence-specific DNA binding activity. More than 90% of p53 missense mutations reside in this region. The C-tenninal domain of p53 protein is responsible for its oligomerization. p53 proteins assemble through this domain to form stable tetramers. The last 28 amino acids of p53 act as a site of negative regulation for the sequence-specific DNA binding function of the central domain (Hupp and Lane, 1994) and may also encode a non-specific DNA binding site (Levine, 1997) Normally the amount of p53 protein in a cell at steady state is low because of its relative short half life (about 20 minutes). Different types of DNA damage can activate p53 protein, including double-strand breaks induced by y-irradiation, ultraviolet irradiation, and chemical damage to DNA. This results in a rapid increase in the level of p53 protein in cells and activation of p53 as a transcription factor (Cox and Lane, 1995). One of the downstream genes of p53 is the p21 protein, an universal inhibitor of cyclin- dependent kinases (El-Deiry et al., 1993). The p21 protein binds to a number of cyclin- 43 cdk complexes, inhibiting cdk kinase activity and blocking cell cycle progression (Xiong et al., 1993). The p21 protein also binds to proliferating cell nuclear antigen (PCNA), blocking its role as a DNA polymerase processitivity factor in DNA replication (Waga et al., 1994). Therefore, p21 can act on cyclin-cdk complexes and PCNA to stop DNA replication, and arrest cells in the G1 phase. This p53-mediated G1 arrest is believed to give cells sufficient time to repair DNA damages before the cells enter into the S phase. The p53 protein has also been shown to mediate GZIM phase arrest. When mitotic spindle inhibitors, such as nocodazole, are added to cells with wild type p53, the cells are arrested in 62. In the absence of p53, these cells will reinitiate DNA synthesis, increasing the ploidy of the cells (Cross et al., 1995). In addition, p53 appears to be involved in regulating the number of centrosomes in a cell. Mouse embryo fibroblasts (MEFs) from p53 null mice produce abnormal numbers of centrosomes after a few doublings in cell culture and initiate spindles with three or four poles, whereas MEFs from normal mice in culture at the same passage level do not exhibit this phenotype (Fukasawa et al., 1996). This p53-mediated GZIM check point may account for the phenotype of genomic instability that is commonly associated with a p53 mutation. Another important function of the p53 protein is to regulate apoptosis. Normal thymocytes will undergo apoptosis in response to DNA damage, whereas thymocytes from p53 null mice do not (Lowe et al., 1993). p53 can also initiate apoptosis in response to the expression of a viral or cellular oncogene. The expression of the adenovirus E1A protein in rat fibroblasts stabilizes and activates p53 protein. The resultant cells die of apoptosis (Debbas and White, 1993). Cells overexpressing E2F-1 and a temperature sensitive mutant p53 protein undergo apoptosis at 32°C but not at 37-39°C, where p53 is inactive (Wu and Levine, 1994). Similarly, cells overexpressing myc and a temperature-sensitive p53 have a temperature-sensitive apoptotic response 44 (Wagner et al., 1994). Besides these factors, hypoxia is able to stimulate p53 levels and induce apoptosis (Graeber et al., 1996). It has been suggested that this process represents another way that p53 may act as a tumor suppressor (Kinzler and Vogelstein, 1996). Most tumors are initiated by genes other than p53 and go through many rounds of clonal expansion to reach a critical size when the blood supply becomes rate-limiting. Angiogenesis is required for clinically apparent growth of a tumor. The resultant hypoxia induces a p53- mediated apoptosis to limit the progression of tumors. In summary, the current knowledge of p53 function suggests that the p53 protein acts to protect the organism from genetic damage. In response to damage or potential damage to the DNA, p53 protein initiates a protective cell cycle arrest or apoptotic cell death. A number of factors affect the decision of a cell to enter a p53-mediated cell cycle arrest or apoptotic pathway. Under conditions in which the DNA is damaged, growth factors are deprived, or an activated oncogene is forcing the cell into proliferation, p53-mediated apoptosis prevails. In this way, cells with damaged DNA or cells in a stressed environment are eliminated in a p53-dependent apoptotic event. Loss of p53 gene function allows the propagation of cells with genetic damage and this may be a key step in the development of neoplasia (Lane, 1994; Levine, 1997). 3.2. Function of the Rb tumor suppressor gene Rb, the product of the retinoblastoma tumor suppressor gene, exerts its cell growth inhibitory function mainly by inhibiting the activity of transcription factors E2Fs. E2Fs are a family of heterodimeric transcription factors that can transactivate genes whose products are important for S phase entry, including most notably c-myc, B-myb, cdc2, thymidine kinase and dihydrofolate reductase (Nevins, 1992; Lathangue, 1994). Rb exerts most of its effect in a defined window of time in the first two thirds of the G1 45 phase of the cell cycle. Cells entering G1 from mitosis require exposure to serum mitogens continuously until several hours before the onset of S phase; thereafter they become relatively serum-independent. This transition from a serum-dependent to serum-independent state is demarcated by a discrete point in time, termed R (restriction) point (Pardee, 1989). Before a cell reaches the R point, the Rb protein is found in an underphosphorylated form. The hypophosphorylated Rb binds to a subset of E2F complexes, converting them to repressors that constrain expression of E2F target genes (Weinberg, 1995; Sherr, 1997). During the last several hours of G1, the Rb protein is initially phosphorylated by cyclin D-dependent kinases and then by cyclin E- CDK2 complex. Phosphorylated Rb protein dissociates from E2Fs, enabling them to transactivate the genes important for S phase entry. A diverse body of evidence indicates that the Rb protein is involved in the pathogenesis of a variety of human tumors. In retinoblastomas, in small cell lung carcinomas, and in many sarcomas and bladder carcinomas, Rb function is lost through mutations of the Rb gene (Horowitz et al., 1990). In the great majority of cervical carcinomas, the cells have been previously infected with one of the oncogenic forms of the papilloma virus and the inactivation of Rb is achieved by the binding of the E7 viral protein (zur Hausen, 1991). As discussed earlier, many tumor cells constitutively overexpress cyclin D1 (see B36), and this in turn activates cyclin D-dependent kinases and phosphorylates Rb protein. Taken together, as a direct consequence of Rb inactivation (either by genetic mutation or by functional inactivation), E2Fs are liberated from Rb inhibition and the progression of cells into late G1 and S phase become uncontrolled. 3.3. Function of the p16 tumor suppressor gene The cell cycle is controlled by the sequential activation and inactivation of cyclin- 46 dependent kinases. As discussed earlier, among the members of the cyclin family, D1 can be an oncogene and functions as an effector of mitogen—induced proliferation acting during the G1 phase of the cell cycle. The D1-dependent kinases are subject to additional levels of regulation, including the association with inhibitory subunits (Morgan, 1995; Sherr and Roberts, 1995). One such inhibitor, p16, specifically binds and inhibits the cyclin D-dependent kinases by competing with D cyclins. Since cyclin D-dependent kinase activity is required to phosphorylate the Rb protein during the middle to late G1 phase as described above, p16 can negatively regulate cell proliferation by suppressing hyperphosphorylation and functional inactivation of the Rb protein. The involvement of p16 in the development of human cancers was implied by the observation that the p16 gene is mutated in many tumor-derived cell lines and maps to chromosome 9p21, a region frequently altered in human malignancies (Kamb et al., 1994; Nobori et al., 1994; Okamoto et al., 1994). Several mechanisms of p16 inactivation have been characterized. Point mutation and small deletions are common in pancreatic adenocarcinomas, esophageal carcinomas, and in families with hereditary susceptibility to melanoma (Hirama and Koeffler, 1995; Pollock et al., 1996). Homozygous deletions of the p16 locus occur commonly in non-small cell lung carcinomas, head and neck tumors, prostate tumors and bladder carcinomas (Hirama and Koeffler, 1995). Finally, methylation of the p16 gene locus is common in breast and colon cancers and results in silencing of the p16 promoter (Herman et al., 1995). Consistent with its tumor suppressor function, p16 null mice spontaneously develop a spectrum of different tumors by 6 months of age, with the rate of tumor formation accelerating in response to carcinogen treatment (Serrano et al., 1996). Cultured p16”' embryo fibroblasts do not enter senescence, and unlike their wild type counterparts, they can be transformed by oncogenic ras alone (Serrano et al., 1996). Presumably, 47 p16 loss might mimic the overexpression of cyclin D1, leading to the hyperphosphorylation and inactivation of Rb protein. D. Carcinogenesis is a multistep process: The role of oncogene and tumor suppressor gene 1. Epidemiological studies of cancer Cancer is predominantly a disease of the elderly, with the risk of acquiring this disease increasing with age. The first clue to the multistep nature of carcinogenesis comes from epidemiological studies. Earlier epidemiological studies of cancer incidence as a function of age using mathematical modeling have shown that for adult human tumors four to six genetic changes were required for genesis of a tumor (Arrnitage and Doll, 1954; Peto, 1977; Dix, 1989). Recently Renan (1993) used the same method but a better defined data set to address the question of the number of mutational changes required for 28 different human malignancies. By plotting the log of the age-specific mortality rate against the log of the age in years of the person affected, and determining the best-fit linear regression coefficients for each tumor type, he concluded that the common adult human cancers of the lip, stomach, liver, pancreas, kidney, skin and bladder required 7 to 8 mutational changes, and tumors with very late onset, such as prostate cancer, required 12 changes. Though this type of studies provides us a general idea about the multistep nature, it can not tell what genetic changes are involved. 2. Experimental animal studies: Mouse skin carcinogenesis A classical model of multistep carcinogenesis is the pathogenesis of mouse skin cancer (Mottram, 1944; Boutwell, 1964; 1974; Hennings et al., 1990; Yuspa, 1994). Sequential application of chemical agents to mouse skin can induce tumors, and tumor development can be divided into three stages: initiation, promotion and progression. Typically, tumor initiation is brought about by the single application of a mutagen, such 48 7,12-dimethylbenz(a)anthracene (DMBA); promotion is carried out by repeated application of a phorbol ester, such as 12-O-tetradecanoylphorboI-13-acetate (T PA) or by a natural promoting stimulus such as wounding. Papillomas begin to appear at 12 to 20 weeks after the promotion has begun and by about 1 year, about 40% to 60% of the animals have some papillomas that become squamous cell carcinomas. If the promoting agents are given alone or before the initiating agent, usually no malignant tumors occur. Detailed studies of the molecular basis of each stage of the skin tumor development indicate that initiation involves a permanent, heritable changes in the gene expression of the initiated cells which produces a subtle change in the keratinocyte phenotype. Genetic analyses revealed that c-Ha-ras alterations are associated with the initiated phenotype. It has been shown that c-Ha-ras mutations are usually heterozygous in papillomas and can be detected in initiated skin prior to the emergence of tumors (Nelson et al., 1992). Furthermore, the initiating agent used determines the existence, nature, and site of the c-Ha-ras mutation (Quintane et al., 1986; Brown et al., 1990). For example, when DMBA is metabolized, the diol epoxide produced primarily binds to adenine residues in DNA (Cheng et al., 1988). When H-ras mutations were analyzed in skin tumors of DMBA-treated mice, the mutations were A181 to T transversions in 45 of 50 tumors (Brown et al., 1990). When a subpopulation of keratinocytes isolated from carcinogen-initiated skin are cultured in vitro, they resist the Ca2+ signal for terminal differentiation and evolve as foci which continue to grow in medium containing >0.1 mM Ca”. Biochemical analyses indicate that overexpression of the TGFa protein and an alteration of PKC are essential for mediating this phenotype. Differential modification of PKC isoforms, particularly activation of PKCa through increased levels of cellular diacylglycerol and functional 49 inhibition of PKC8 by tyrosine kinase, produces keratinocytes with enhanced proliferative capacity and reduced sensitivity to signals for terminal differentiation (Yuspa, 1994). Application of tumor promoters to initiated epidermis causes the selective clonal outgrth of initiated cells to produce multiple benign squamous cell papillomas, each representing an expanded clone of initiated cells (Deamant et al., 1987; lannaccone et al., 1987). The mechanisms of exogenous promotions are likely to be epigenetic in most cases because (1) a single genetic change in normal keratinocytes is sufficient to produce a papilloma phenotype (Greenhalgh et al., 1993) and (2) most promoting agents are not mutagens (Yuspa and Dlugosz, 1991). Progression of a papilloma to a carcinoma in mouse skin is usually a spontaneous process. Progression and malignant conversion can be enhanced and accelerated by exposing animals beefing papillomas to a mutagen, supporting a genetic basis for progression (O’Connell et al., 1986; Hennings et al., 1990). Genetic studies indicate that non-random, sequential chromosomal aberrations are associated with progression of mouse skin papillomas; particularly prominent are trisomies of chromosomes 6 and 7 (Yuspa, 1994). Changes in two cellular genes, c-Ha-ras and p53, have been closely identified with malignant conversion of skin tumors. The mutated c-Ha-ras gene, which is heterozygous in papillomas, is frequently homozygous in carcinomas (Bianchi et al., 1990). Mutations in the p53 tumor suppressor gene are rarely found in chemically induced papillomas but are frequently detected in squamous carcinomas, particularly those induced by benzo(a)pyrene (Ruggeri et al., 1991; 1993; Kress et al., 1992). Recent studies indicate that 90% of squamous carcinomas are devoid of TGFB1 and TGFBZ. Direct evidence linking TGF81 loss and accelerated malignant tumor progression comes from studies using keratinocytes cultured from TGFB1 null mice. 50 Introduction of v-Ha-ras oncogene into cultured keratinocytes of TGFB1 null mice or wild-type and heterozygous Iittermates results in papillomas when these cells are grafted to nude mice. However, carcinomas only develop in the TGFB1 null papillomas but not in those of other genotypes (Yuspa, 1994). These results indicate that the TGFB family of growth inhibitors can serve as suppressor of malignant progression. The mouse skin carcinogenesis model clearly demonstrates that the genesis of skin tumors requires non-genetic changes as well as a series of genetic changes. Mutations in oncogenes, e.g., c-Ha-ras, tumor suppressor genes, e.g., p53, altered expression of TGFj31, altered activity of PKC and characteristic chromosomal abnormalities all contribute to the development of the squamous cell carcinomas. 3. Human cancer studies: Colorectal carcinogenesis Colorectal carcinomas have proven to be an excellent model system in which to study the genetic changes involved in the initiation and progression of human solid tumors. There is a well-defined progression from benign to malignant tumors, and the cancers that arise are clonal in origin. Tumors of various stages of dysplasia and malignancy, ranging from benign adenomas to invasive carcinomas, can be obtained surgically. In addition, colorectal cancer exists in both sporadic and inherited form. For example, in patients with familial adenoma polyposis (FAP), hundreds to thousands of adenomatous polyps will develop in their colons and rectums, and a small percentage of these go on to become malignant. The high frequency of polyps at various stages of progression allows one to study the stage-wise pattern of colon cancer. Studies of this inherited cancer syndrome and large number of sporadic tumors allowed Vogelstein and colleagues to identify the critical genetic events driving colorectal carcinogenesis (Vogelstein et al., 1988; Fearon and Vogelstein, 1990). An early change that occurs in the genome of cells of small, early adenomas from FAP 51 patients and patients without familial predisposition is chromosomal allelic loss of 5q. Linkage analysis and molecular cloning have identified the affected gene on chromosome 5q21 in FAP and the gene is called APC ( for adenomatous polyposis coli) (Groden et al., 1991; Kinzler et al., 1991). The APC gene is mutated in the gennline of FAP patients (Mandi et al., 1994) while truncating APC mutations have been identified in 60% of sporadic colorectal cancers and adenomas (Powell et al., 1992; lchii et al., 1993). Both copies of APC are inactivated either by mutations on each allele, or by a mutation on one allele and a structural deletion in the other. The data indicate that APC is a tumor suppressor gene, and one or both alleles of the APC gene are lost orinactivated at an early stage in the development of colorectal cancer. Mutations in the K-ras oncogene have been identified in about 10% of small adenomas, 50% of larger adenomas and in 50% of carcinomas. Such ras mutations, usually in codons 12 or 13, tend to be observed in more dysplastic adenomas. Ras gene mutations appear to occur in one cell of a preexisting adenoma and confers it a growth advantage. Through clonal expansion of the cells with the mutation, a small adenoma is converted into a larger and more dysplastic one. Loss of heterozygosity of additional tumor suppressor genes appears to be critical in later stages of colorectal tumorigenesis. The chromosomes most frequently deleted include chromosome 18q and 17p. 18q is deleted in 50% of late adenomas and more than 70% of carcinomas (Vogelstein et al., 1988). A candidate tumor suppressor gene from this region has been identified. This gene, termed DCC (for deleted in colorectal cancer), encodes a protein with putative cell adhesion properties. Its expression is absent or reduced in colorectal carcinomas, suggesting that its involvement in normal cell-cell or cell-matrix interactions is required to maintain a normal state of differentiation. The loss of a large portion of one copy of chromosome 17p has been 52 observed in more than 75% of carcinomas. This loss is rarely seen in adenomas at any stage. The common region lost on 17p contains the p53 tumor suppressor gene. Nucleotide sequencing analysis of the p53 cDNA derived from colorectal cancers has shown that in 70% to 80% of the cases there is a missense mutation in the remaining p53 allele in the cancer cells. Additional chromosome losses, including 1q, 4p, 6q, 8p, 9g and 22q, has been observed in colorectal cancers. On average, colorectal carcinomas contain four or five allelic losses. Patients with more than the median number of losses in their tumors have a poorer prognosis. In summary, colorectal tumors appear to arise as a result of the mutational activation of specific oncogenes and inactivation of specific tumor suppressor genes. Mutations in at least four to five genes are required for the formation of a malignant tumor. Fewer changes are sufficient for benign tumorigenesis. These studies clearly demonstrate the complex multistep nature of human tumor development. 4. Transformation of human cells in culture One of the advantages in using human cells in culture to study neoplastic transformation is that it provides a means to dissect the carcinogenesis process under well-defined conditions. However, normal human cells in culture have never been found to undergo spontaneous malignant transformation and have proven to be extremely difficult to be malignantly transformed by any means (McCormick and Maher, 1988; Rhim, 1993). Recently neoplastic transformation of human cells in culture has been achieved in a stepwise fashion-immortalization and malignant conversion of the immortalized cells. These studies support a multistep process for neoplastic transformation and provide insights into the molecular mechanisms underlying the process. 4.1. Immortalization of human cells in culture 53 Normal human cells in culture have a limited life span, beyond which the cells enter the terminally nondividing state referred to as senescence (Hayflick and Moorhead, 1961). Finite life span normal human cells can only undergo two successive clonal selections before they enter crisis and senesce (McCormick and Maher, 1988). Earlier studies trying to transform finite life-span human cells in culture were not successful, suggesting that acquiring immortality is a prerequisite if a cell is to acquire sequentially all the genetic changes needed to become malignant (Kuroki and Huh, 1993; McCormick and Maher, 1994). Whether this is the case for cells in the human body is not known for certain. What is known is that many malignant tumor-derived cells can be grown indefinitely in culture, whereas cells from normal tissues are never able to grow indefinitely in culture. Human cells can be immortalized by repeated treatment with chemical and physical carcinogens, and by infection or transfection with certain viral genes. However it occurs at a very low frequency (McCormick and Maher, 1988; Shay et al., 1991; Bai et al., 1993; Kuroki and Huh, 1993). In order to immortalize human fibroblasts, for example, Namba et al. (1988) had to expose them to more than 10 treatments with either 4-nitroquinoline 1-oxide or gamma-irradiation and has been successful only a few times. Nevertheless, immortalization of human epithelial cells, for example, epidermal keratinocytes, bronchial epithelial cells, mammary epithelial cells, and prostate epithelial cells, have been achieved by infection with the AD12-SV40 hybrid virus or human papilloma virus (Kuroki and Huh, 1993; Rhim, 1993). Immortalization of diploid human foreskin fibroblast has been achieved in this laboratory by transfection with a v-myc oncogene. An early passage, foreskin-derived normal human fibroblast cell line was transfected with a plasmid carrying the neo gene and a v- myc gene. The transfectants were selected for geneticin resistance and clonally—derived 54 cell strains expressing the v-myc protein were isolated and propagated for many generations. Eventually all cell strains senesced, but among the senescing progeny viable cells could be seen. These eventually gave rise to an infinite life span cell strain designated MSU-1.1 (Morgan et al., 1991). This experiment demonstrates that expression of the v-myc oncoprotein alone is not sufficient to cause human fibroblasts to acquire an infinite life span, but we do not yet know what additional genetic change(s) is required. At present the mechanisms underlying immortalization are poorly understood. Careful analyses of the immortalization process suggest that immortalization is caused by multiple genetic changes. For example, following infection by SV40 virus or stable transfection with the SV40 T-antigen, normal proliferating human fibroblasts replicate for about 20-30 more population doublings beyond their normal senescent point. The population then enters a state of crisis, during which cell number remains constant or declines due to an increase in cell death. From this crisis population arise the rare, immortal clones, presumably due to additional genetic events (Wright and Shay, 1992). It has been reported that (Shay and Wright, 1989) for a population of SV40 T-antigen transfected human lung fibroblasts, the frequency of immortalization is about 3 X 10‘7. Somatic cell hybridization between an immortal cell line and a finite life span cell line results in cells with limited life span, indicating that immortalization is caused by recessive gene mutations (Smith and Pereira-Smith, 1996). Recently, a number of studies indicate that the p53 and Rb tumor suppressor genes may play a causal role in immortalization of certain cell types (Shay et al., 1991; Vojta and Barrett, 1995). For example, fibroblasts from seven out of eight Li-Fraumeni syndrome patients escaped senescence and were immortalized spontaneously in culture (Bischoff et al., 1990). Hara et al. (1991) reported that targeted functional knock-out of the Rb and p53 with 55 antisense oligomers resulted in an extended life span of human fibroblasts. 4.2. Neoplastic transformation of Immortal human cells While immortality is not sufficient for transformation, most immortalized cells have an increased sensitivity for spontaneous, carcinogen- or oncogene-induced neoplastic progression. For example, ADIZ-SV40 hybrid virus immortalized human epidermal keratinocytes can be malignantly transformed by retroviral oncogenes such as H-ras, fms, erbB and src; they can also be transformed by chemical carcinogens or by x-ray irradiation treatment (Reviewed by Rhim, 1993). This laboratory has successfully converted MSU-1.1 human fibroblasts into malignant cells by transfection of an activated H-ras or an N-ras oncogene in a vector engineered for overexpression of the oncogene. Expression of the same ras oncogenes at the level found for the endogenous H-ras or N-ras proto-oncogene did not cause malignant transformation. However, if MSU-1.1 cells that expressed a transfected H-ras or v-sis oncogene at low levels were subsequently transformed with a v-fes oncogene, then the cells became malignant (Lin et al., 1995). These studies indicate that more than one genetic changes are needed for the malignant transformation of MSU-1.1 cells. They also demonstrate the complementary role between oncogenes. In summary, normal human cells in culture can be immortalized by a variety of means (viruses, chemical carcinogens, irradiation and oncogene transfection). Additional exposure of these immortal cells to a carcinogenic agent and appropriate selection can result in malignantly transformed cells. Thus these studies demonstrate that similar to tumor development in vivo, neoplastic transformation of human cells in culture is indeed a multistep process. LIST OF REFERENCES LIST OF REFERENCES Aaronson, SA. (1991). Growth factors and cancer. Science, 254:1146-1153. Abbadie, C., Kabrun, N., Bouali, F et al. (1993). High levels of c-rel expression are associated with programmed cell death in the developing avian embryo and in bone marrow cells in vitro. Cell, 75:899-912. Abrahams, P.J., Mulder, C., van der voode A, et al.. (1975) Transformation of primary rat kidney cells by fragments of simian virus 40. J. Virol., 16:818-823. Albanese, C., Johnson, J., Watanabe, G., Eklund, N., Vu, 0., Arnold, A., and Pestell, R. G. (1995). Transforming p21ras mutants and c-Ets-2 activate the cyclin D1 promoter through distinguishable regions. J. Biol. Chem, 270: 23589-23597. Alonso, G., Koegl, M., Mazurenko, N., and Courtneidge, SA. (1995). Sequence requirements for binding of Src family tyrosine kinases to activated growth factor receptors. J. Biol. Chem., 279:9840-9848. Ames, B.N., Durston, W.E., Yamasaki, E., and Lee, FD. (1973). Carcinogens are mutagens: simple test system combining liver bacteria for detection. Proc. Natl. Acad. Sci. USA., 70:2281-2285. Angel, P., and Karin, M. (1991). The role of Jun, Fos and AP-1 complex in cell- proliferation and transformation. Biochim. Biophys. Acta, 1072:129-157. Annitage, P., and Doll, R. (1954). The age distribution of cancer and a multistage theory of carcinogenesis. Brit. J. Cancer Res., 8:1-12. Bai, L., Mihara, K., Kondo, Y., Honma, M., and Numba, M. (1993). Immortalization of normal human fibroblasts by treatment with 4-nitroquinoline 1-oxide. Int. J. Cancer, 53:451-456. - Balaban, G., Gilbert, F., Nichols, W., et al.., (1982). Abnormalities of chromosome 13 in retinoblastomas from individuals with normal constitutional karyotypes. Cancer Genet. Cytogenet, 6:213-221. Bassin, RH, and Noda, M. (1987). Oncogene inhibition by cellular genes. Adv. Viral. Oncol. 6, 103-127. 56 57 Bernhard, W. (1960). The detection and study of tumor viruses with the electron microscope. Cancer Res., 20:712-727. Berridge, M.J., and Irvine, RF. (1989). lnositol phosphates and cell signaling. Nature, 341:197-205. Bianchi, A.B., Aldaz, CM, and Conti, C.J. (1990). Nonrandom duplication of the chromosome bearing a mutated Ha-ras-1 allele in mouse skin tumors. Proc. Natl. Acad. Sci. USA., 87:6902-6906. Birch, J.M. (1994). Familiar cancer syndromes and clusters. Brit. Med. Bullet, 50:624- 639. Bischoff, F.Z., Yim, S.O., Pathak, S., Grant, 6., Siciliano, M. J., Giovanella, B. C., Strong, L. C., Tainsky, M. A. (1990). Spontaneous abnormalities in normal fibroblasts from patients with Li-Fraumeni cancer syndrome: aneuploidy and immortalization. Cancer Res. 50: 7979-7984. Bishop, J.M. (1991) Molecular themes in oncogenesis. Cell, 64:235-248. Bishop, J. M. (1987) The molecular genetics of cancer. Science, 235:305-311. Bishop, J.M. (1985). Wral oncogenes. Cell, 42:23-38. Bishop, J.M., and Varmus, H. Functions and origins of retroviral transforming genes. In Weiss, W., Teich, N., Varmus, H., and Coffin, J. (Eds): RNA tumor viruses. Cold Spring Harbor, NY, Cold Spring Harbor Laboratory Press, 1982, pp.999-1108. Bissonnette, R.P., Echeveri, F., Mahboubi, A., and Green, D. (1992). Apoptotic cell death induced by c-myc is inhibited by bcl-2. Nature, 359:552-554. Bodrug, S.E., Warner, B.J., Bath, M.L., Lindeman, G.J., Harris, A.W., and Adams, J.M. (1994). Cyclin D1 transgene impedes lymphocyte maturation and collaborates in lymphomagenesis with the myc gene. EMBO J., 13:2124-2130. Bolden, J.B., Rowley, R.B., Spana, C., and Tsygankov, A.Y. (1992). The Src family of tyrosine protein kinases in hemopoietic signal transduction. FASEB J. 6:3403-3409. Bos, J.L. (1989). ras oncogene in human cancer: A review. Cancer Res. 49:4682- 4689. Boume, H.R., Sanders, DA, and McCormick, F. (1990). The GTPase superfamily: l. A conserved switch for diverse cell functions. Nature, 348:125-131. Boutwell, R.K. (1974). The function and mechanism of promoters of carcinogenesis. CRC Crit. Rev. Toxicol., 2(4): 419-43. Boutwell, R.K. (1964). Some biological aspects of skin carcinogenesis. Prog. Exp. Tumor Res., 4: 207-250. S8 Boveri, T. (1929). The origin of malignant tumors. (Baltimore: The Vlfllliams and Wilkins, 00.), pp.26—27. Bronner, M.P., Culin, C., Reed, J., and Furth, E. (1995). BcI-2 proto-oncogene and the gastrointestinal mucosal epithelial tumor progression model. Am. J. Pathol., 146:20-26. Bronner, C.E., Baker, S.M., Morrison, P.T., Warren, G., Smith, L.G., Lescoe, M.K., Kane, M., Earabino, C., Lipford, J., Lindblom, A, et al.. (1994). Mutation in the DNA mismatch repair gene homologue hMLH1 is associated with hereditary non-polyposis colon cancer. Nature, 368: 258-261. Brott, B.K., Decher, S., Shafer, J. et al., (1991). GTPase-activating protein (GAP) interacts with viral and cellular src kinase. Proc. Natl. Acad. Sci. USA, 88:755-759. Brown, MT, and Cooper, J.A. (1996). Regulation, substrates and regulation of src. Biochim. Biophys. Acta, 1287:121-149. Brown, K., Buchmann, A, and Balmain, A. (1990). Carcinogen-induced mutations in the mouse c-Ha-ras gene provide evidence of multiple pathways for tumor progression. Proc. Natl. Acad. Sci. USA, 87: 538-542. Cabilly-Snyder, L., Yang-Feng, T., Francke, U., and George, D.L. (1987). Molecular analysis and chromosomal mapping of amplified gene isolated from a transformed mouse 3T3 cell lines. Somat. Cell Mol. Genet, 13:235-244. Cacace, A.M., Guadagno-Nichols, S., Krauss, R., et al., (1993). The epsilon isoform of PKC is an oncogene when overexpressed in rat fibroblasts. Oncogene, 8:2095-2104. Carter, R., Cosenza, S.C., Pena, A, Lipson, K., Soprano, DR, and Soprano, K. J. (1991). A potential role for c-jun in cell cycle progression through late G1 and S. Oncogene, 6: 229-235. Chan, A.M., Chedid, M., McGovern, E.S., Popescu, N.C., Miki, T., and Aaronson, SA (1993). Expression cDNA cloning of a serine kinase transforming gene. Oncogene, 8: 1329-33. Chan, A.M., Miki, T., Meyers, K.A., and Aaronson, SA. (1994). A human oncogene of the RAS superfamily unmasked by expression cDNA cloning. Proc. Natl. Acad. Sci. USA, 91: 7558-62. Chang, H.W., Aoki, M., Fruman, D., Auger, K.R., Bellacosa, A, Tsichlis, P.N., Cantley, L.C., Roberts, TM, and Vogt, PK. (1997). Transformation of chicken cells by the gene encoding the catalytic subunit of PI 3-kinase. Science, 276:1848-1850. Cheng, S.C., Prakash, A. S., Pigott, M. A, Hilton, B. D., Lee, H., Harvey, R. G., and Dipple, A. (1988). A metabolite of the carcinogen 7,12-dimethylbenz[a]anthracene that reacts predominantly with adenine residues in DNA. Carcinogenesis, 9: 1721-1723. Cleary, ML. and Sklar, J. (1985). Nucleotide sequence of a t(14;18) chromosome 59 breakpoint in follicular lymphoma and demonstration of a breakpoint-cluster region near a transcriptionally active locus on chromosome 18. Proc. Natl. Acad. Sci. USA, 82:7439-7443. Cleary, M.L., Smith, SD, and Sklar, J. (1986). Cloning and structural analysis of cDNA for bcI-2 and a hybrid bcI-2/immunoglobulin transcript resulting from the t(14;18) translocation. Cell, 47:19-28. Cleaver, J.E., and Kraemer, K.H. (1989). In The Metabolic Basis of Inherited Disease. 6th Edition, Scriver,C.A, Beaudet, A.L., Sly, W.S., and Vale, D. Eds. New York, McGraw-Hill, pp. 2949-2971. Cleaver, J.E. (1990). Do we know the cause of xeroderma pigmentosum? Carcinogenesis, 1 1 :875-882. Cochet, C.C., Filhol, O., Payrastre, B., Hunter, T., and Gill, GM. (1991). Interaction between the epidermal growth factor receptor and phosphoinositide kinases. J. Biol. Chem., 266:637-644. Cochran, B.H., Zumstein, P., Zullo, J., Rollins, B., Mercola, M., and Stiles, CD. (1987). Differential colony hybridization: molecular cloning from a zero data base. Methods Enzymol., 147: 64-85. Collins, PS. (1991). Of needles and haystacks: Finding human disease genes by positional cloning. Clin. Res., 39:615-623. Conney, AH. (1982). Induction of microsomal enzymes by foreign chemicals and carcinogenesis by polycyclic aromatic hydrocarbons. Cancer Res., 4224875. Cook, S.J., Rubinfeld, B., Albert, 1., and McCormick, F. (1993). RapV12 antagonizes ras-dependent activation of ERK1 and ERK2 by LPA and EGF in Rat-1 fibroblasts. EMBO J.,12: 3475-3485. Cox, LS, and Lane, DP. (1995). Tumour suppressors, kinases and clamps: how p53 regulates the cell cycle in response to DNA damage. Bioessays, 17: 501-508. Crews, CM, and Erikson, R.L. (1993). Extracellular signals and reversible protein phosphorylation: What to Mek of it all. Cell, 74:215-217. Cross, M., and Dexter, TM. (1991). Growth factors in development, transformation, and tumorigenesis. Cell, 64:271-280. Cross, S.M., Sanchez, C.A., Morgan, C.A., Schimke, M.K., Ramel, S., ldzerda, R.L., Rasking, W.H., and Reid, B.J. (1995). A p53-dependent mouse spindle check point. Science, 267: 1 353-1 356. Curran, T., Peters, G., Van, B.C., Teich, NM, and Verma, 1M. (1982). FBJ murine osteosarcoma virus: identification and molecular cloning of biologically active proviral DNA. J. \firol.,44:674-682. 60 Dal-Cin, P., and Sandberg, AA (1989). Chromosomal aspects of human oncogenesis. Crit. Rev. Oncog., 1: 113-26. Dalla-Favera, R., Martinotti, S., Gallo, R., Erikson, J., and Croce.C.M. (1983). Translocation and rearrangement of the c-myc oncogene locus in human undifferentiated B-cell lymphomas. Science, 219:963-967. Datson, N.A, van-de Vosse, E., Dauwerse, H.G., Bout, M., van-Ommen, G.J., and den-Dunnen, J.T. (1996). Scanning for genes in large genomic regions: cosmid-based exon trapping of multiple exons in a single product. Nucleic Acids Res., 24: 1105-1111. De Mars, R. 23rd Annual Symposium, Fundamental Cancer Research 1969. Baltimore, Williams and Wilkins 1970; pp. 105-106. De Klein, A, and Hagemeijier, A. (1984). Cytogenetics and molecular analysis of the Ph’ translocation in chronic myeloid leukemia. Cancer Surv., 3:515. Deamant, F.D., and lannaccone, PM. (1987). Clonal origin of chemically induced papillomas: separate analysis of epidermal and dermal components. J. Cell Biol., 88:305-312. Debbas, M., and White, E. (1993). Wild-type p53 mediates apoptosis by E1A which is inhibited by E1B. Genes Dev., 7:546-554. DeKlein, A, van Kessel, A.G., Grosveld, G., Bartram, C.R., Hagemeijer, A, Bootsma, D., Spurr, N.K., Heisterkamp, N., Grotten, J., and Stephenson, JR (1982). A cellular oncogene is translocated to the Philadelphia chromosome in chronic myelocytic leukemia. Nature, 300:765-767. DeIIi-Bovi, P., Curatola, AM., Kern, F.G., Greco, A, lttmann, M., and Basilico, C. (1987). An oncogene isolated by transfection of Kaposi's sarcoma DNA encodes a growth factor that is a member of the FGF family. Cell, 50: 729-737. DePinho, R., Mitsock, L., Hatton, K., Ferrier, P., Zimmerman, K., Legouy, E., Tesfaye, A, Collum, R., Yancopoulos, G., Nisen, P., et al. (1987). Myc family of cellular oncogenes. J. Cell Biochem., 33: 257-66. Der, C.J., Krontiris, T.G., and Cooper, GM. (1982). Transforming genes of human bladder and lung carcinoma cell lines are homologous to the ras genes of Harvey and Kirsten sarcoma viruses. Proc. Natl. Acad. Sci. USA, 79:3637. Dix, D. (1989). The role of aging in cancer incidence: an epidemiological study. J. Gerontol., 44:10-18. Doolittle, R.F., Hunkapiller, M.W., Hood, L.E., Devare, S.G., Robbins, K.C., Aaronson, SA and Antoniades, HM (1983). Simian sarcoma virus onc gene, v-sis, is derived from the gene(s) encoding a platelet-derived growth factor. Science, 221:275—277. Dryja, T., Rapaport, J., Joyce, J., et al.. (1986). Molecular detection of deletions 61 involving band q14 of chromosome 13 in retinoblastomas. Proc. Natl. Acad. Sci. USA, 83:7391-7394. Duyk, G.M., Kim, S.W., Myers, RM, and Cox, DR. (1990). Exon trapping: a genetic screen to identify candidate transcribed sequences in cloned mammalian genomic DNA. Proc. Natl. Acad. Sci. USA, 87: 8995-8999. Egan, SE, and Weinberg, RA (1993). The pathway to signal achievement. Nature, 365:781-783. Egan, S.E., Giddings, B.W., Brooks, M.W., Buday, L., Sizeland, AM., and Weinberg, RA (1993). Association of $05 ras exchange protein with Grb2 is implicated in tyrosine kinase signal transduction and transformation. Nature, 363245-51. EI-Deiry, W.S., Tokino, T., Velculescu, V.E., Levy, D.B., Parsons, R., Trent, J.M., Lin, 0., Mercer, W.E., Kinzler, K.W., and Vogelstein, B. (1993). WAF1, a potential mediator of p53 tumor suppression. Cell, 75:817-825. Erikson, J., ar-Rushdi, A, Drwinga, H.L., Nowell, PC, and Croce, CM. (1983). Transcriptional activation of the c-myc oncogene in Burkitt's lymphoma. Proc. Natl. Acad. Sci. USA, 80:820-824. Evan, G.l., Wyllie, AH., Gilbert, C.S. et al.. (1992). Induction of apoptosis in fibroblasts by c-myc protein. Cell, 69:119-128. Evans, E.P., Burtenshaw, M.D., Brown, B.B., Hennion, R., and Harris, H. (1982). The analysis of malignancy by cell fusion. IX. Reexamination of clarification of the cytogenetic problem. J. Cell Sci., 56:113-130. Fakhaizadeh, S.S., Trusko, SP, and George, D.L. (1991). Tumorigenic potential associated with enhanced expression of a gene that is amplified in a mouse tumor cell line. EMBO J., 10:1565-1569. Fanidi, A, Harrington, EA, and Even. G. (1992). Cooperative interaction between c- myc and bcl-2 protooncogenes. Nature, 359:554-556. Fearon, ER, and Vogelstein, B. (1990). A genetic model for colorectal tumorigenesis. Cell, 61:759-767. Fearon, ER, and Jones, PA (1992). Progressing toward a molecular description of colorectal cancer development. FASEB J., 6:2783-2790. Feig, LA (1993). Science, 260:767-768. Femandez-Pol, J.A., Klos, DJ, and Hamilton, PD. (1993). A growth factor-inducible gene encodes a novel nuclear protein with zinc finger structure. J. Biol. Chem, 268: 21198-21204. Fishel, R., Lescoe, M.C., Rao, M.R., Copeland, N.G., Jenkins, N.A., Garber, J., Kane, 62 M., et al., (1993). The mutation mutation gene homologue MSH2 and its association with hereditary nonpolyposis colon cancer. Cell, 75:1027-1037. Forrest, D., and Curran, T. (1992). Crossed signals: oncogenic transcription factors. Curr. Opin. Genet. Dev., 2: 19-27. Francke, U. (1978). Retinoblastoma and chromosome 13. Birth Defects, 12:131-137. Frei, J. V., Swenson, D. H., Warren, W., and Lawley, P. D. (1978). Alkylation of deoxyribonucleic acid in vivo in various organs of CS7BL mice by the carcinogens N-methyl-N-nitrosourea, N-ethyl-N-nitrosourea and ethyl methanesulphonate in relation to induction of thymic lymphoma. Some applications of high-pressure liquid chromatography. Biochem. J., 174: 1031-1044. Friend, S.H., Bemards, R., Rogelj, S., Weinberg, R.A, Rapaport, J.M., Albert, OM, and Dryja, TR (1986). A human DNA segment with properties of the gene that predisposes to retinoblastomas and osteosarcoma. Nature, 323:643-646. Friis, R. R., Toyoshima, K., and Vogt, P. K. (1971).Conditional lethal mutants of avian sarcoma viruses. 1. Physiology of ts 75 and ts 149. Virology. 43: 375-389. Fukasawa, K., Choi, T., Kuriyama, R., Rulong, S., and Vande Woude, GE (1996). Abnormal centrosome amplification in the absence of p53. Science, 271:1744-1747. Galaktionov, K., Jessus, C., and Beach, D. (1995). Raf1 interaction with Cdc25 phosphatase ties mitogenic signal transduction to cell cycle activation. Genes Dev., 9: 1046-1058. Gallego, C., Gupta, S.K., Heasley, L. E., Qian, N. X., and Johnson, G.L. (1992). Mitogen-activated protein kinase activation resulting from selective oncogene expression in NIH 3T3 and rat 1a cells. Proc. Natl. Acad. Sci. USA, 89: 7355-7359. Gallie, B.L., Squire, J.A., Goddard, A, et al.., (1990). Biology of disease: Mechanisms of oncogenesis in retinoblastomas. Lab Invest, 62:394-408. Garber, J.E., Goldstein, AM., Kantor, A.F., Dreyfus, M.G., Fraumeni, J.F., and Li, F-P. (1991). Follow-up study of twenty-four families with Li-Fraumeni syndrome. Cancer Res., 5126094. Golde, A. (1970). Radio-induced mutants of the Schmidt-Ruppin strain of rous sarcoma virus. Virology, 40: 1022-1029. Goldfarb, M., Shimizu, K., Prucho, M., and Wigner, M. (1982). Isolation and preliminary characterization of a human transforming gene from T24 bladder carcinoma cells. Nature, 296:404-409. Graeber, T.G., Osmanian, C., Jacks, T., Housman, D.E., Koch, C.J., Lowe, S.W., and Giaccia, A.J. (1996). Hypoxia-mediated selection of cells with diminished apoptotic potential in solid tumors. Nature, 379:88-91. 63 Grandis, JR, and Tweardy, DJ. (1993). Elevated levels of transforming growth factor alpha and epidermal growth factor receptor messenger RNA are early markers of carcinogenesis in head and neck cancer. Cancer Res., 53:3579—3584. Graninger, W.B., Seto, M., Boutain, B., Goldman, P., and Korsmeyer, SJ. (1987). Expression of Bcl-2 and Bcl-2-lg fusion transcripts in normal and neoplastic cells. J.Clin. Invest. 8021512. Green, R.S., Lieb, M.E., Weintraub, AS., Gacheru, S. N., Rosenfield, C.L., Shah, S., Kagan, HM, and Taubman, MB. (1995). Identification of lysyl oxidase and other platelet-derived growth factor-inducible genes in vascular smooth muscle cells by differential screening. Lab Invest, 73: 476-482. Greenhalgh, D.A., Rothnagel, J.A., Quintanilla, M.I., Orengo, C.C., Gagne, T.A, Bundman, D.S., Longley, MA, and R00p, DR. (1993). Induction of epidermal hyperplasia, hyperkeratosis, and papillomas in transgenic mice by a targeted v-Ha-ras oncogene. Moi. Carcinogenesis, 7: 99-1 10. Groden, J., Thliveris, A, Samowitz, W., Carlson, M., Gelbert, L., Albertsen, H., Joslyn, 6., Stevens, J., Spirio, L., Robertson, M., et al. (1991). Identification and characterization of the familial adenomatous polyposis coll gene. Cell, 66: 589-600. Gutkind, S.J., Lacal, RM, and Robbim, KC. (1990). Thrombin-dependent association of phosphatidylinositol-3 kinase with p60c-src and p59fyn in human platelets. Mol. Cell Biol., 10:3806-3809. Haefner, B., Baxter, R., Fincham, V.J., Downs, GP, and Frame, MC. (1995). J. Biol. Chem., 270:7937-7943. Hall, M., and Peters, G. (1996). Genetic alterations of cyclins, cyclin-dependent kinases, and Cdk inhibitors in human cancer. Adv. Cancer Res., 68: 67-108. Hansen, M., and Cavenee, W. (1987). Genetics of cancer disposition. Cancer Res., 47:5518-5527. Hara, E., Tsurui, H., Shinozaki, A, Nakada, S., and Oda, K. (1991). Cooperative effect of antisense-Rb and antisense-p53 oligomers on the extension of life span in human diploid fibroblasts, TlG-1. Biochem. Biophys. Res. Commun., 179: 528-534. Harris, H. (1988). The analysis of malignancy by cell fusion: The position in 1988. Cancer Res., 48:3302-3306. Harris, H., Miller, O.J., Klein, G., Worst, P., and Tachibanaq, T. (1969). Suppression of malignancy by cell fusion. Nature, 223:363-368. Harris, H., and Klein, G. (1969). Malignancy of somatic cell hybrids. Nature, 224:1314- 1316. Hartwell, L.H., and Kastan, MB. (1994). Cell cycle control and cancer. Science, 266: 64 1821 -8. Hayflick, L., and Moorhead, P. (1961). The serial cultivation of human diploid cell strains. Exp. Cell Res., 25:585. Helm, S., and Mitelman, F. (1989). Primary chromosome abnormalities in human neoplasia. Adv. Cancer Res., 5221-43. Heisterkamp, N., Stam, K., Grotten, J., deKlein, A. And Grosveld, G. (1985). Structural organization of the bcr gene and its role in Ph' translocation. Nature, 315:758-761. Heldin, CH, and Westermark, B. (1989). Platelet-derived growth factors: a family of isoforms that bind to two distinct receptors. Brit. Med. Bull., 452453-464. Hemminki, A, Tomlinson, l., Markie, D., Jarvinen, H., Sistonen, P., Bjorkqvist, A-M., Knuutila, S., Salovaara, R., Bodmer, W., Shibata, D., Chapelle, A, and Aaltonen, LA (1997). Localization of a susceptibility locus for Peutz-Jeghers syndrome to 19p using comparative genomic hybridization and targeted linkage analysis. Nature Genet, 15:87-90. Hennings, H., Glick, A.B., Greenhalgh, D.A., Morgan, D.L., Strickland, J.E., and Yuspa, SH. (1993). Critical aspects of initiation, promotion, and progression in multistage epidermal carcinogenesis. Proc. Soc. Exp. Biol. Med, 20221-8. Hennings, H., Shores, R.A., Poitier, M.C., Reed, E., Tarone, RE, and Yuspa, SH. (1990). Enhanced malignant conversion of benign mouse skin tumors by cisplatin. J. Natl. Cancer Inst, 82:832-840. Herman, J.G., Merlo, A, Mac, L., Lapidus, R.G., lssa, J-P., Davidson, N.E., Sidransky, D., and Baylin, SB. (1995). Inactivation of the CDKN2/p16IMTS1 gene is frequently associated with aberrant DNA methylation in all common human cancers. Cancer Res., 55:4525-4530. Hill, M., and Hillova, J. (1972). Virus recovery in chicken cells tested Rous sarcoma cell DNA. Nature New Biol., 237235-39. Hirama, T., and Koeffler, HP. (1995). Role of the cyclin-dependent kinase inhibitors in the development of cancer. Blood, 86:841-854. Hockenbery, OM. (1994). Bel-2 in cancer, development and apoptosis. J. Cell Sci., 18:51-55. Housey, G.M., Johnson, MD, Hsiao, W.L. et al., (1988). Overproduction of protein kinase C causes disordered growth control in rat fibroblasts. Cell, 52:343-354. Hollstein, M., Rice, K., Greenblatt, M.S., Soussi, T., Fuchs, R., Sorlie, T., Hovig, E., Smith-Sorensen, B., Montesano, R., and Harris, CC. (1994). Database of p53 gene somatic mutations in human tumors and cell lines. Nucleic Acids Res., 22: 3551-3555. 65 Holt, J.T., Gopal, T.V., Moulton, AD., and Nienhuis, AW. (1986). Inducible production of c-fos antisense RNA inhibits 3T3 cell proliferation. Proc. Natl. Acad. Sci. USA, 83: 4794-4798. Horowitz, J.M., Park, S.H., Bogenmann, E., Cheng, J.C., Yandell, D.W., kaye, F.J., Minna, J.D., Dryja, T.P., and Weinberg, RA (1990). Frequent inactivation of the retinoblastoma antioncogene is restricted to a subset of human tumor cells. Proc. Natl. Acad. Sci. USA, 87:2775-2779. Howe, LR, and Marshall, C.J. (1993). Lysophosphatidic acid stimulates mitogen-activated protein kinase activation via a G-protein-coupled pathway requiring p21ras and p74raf-1. J. Biol. Chem., 268: 20717-20720. Hsiao, W.L., Housey, G.M., Johnson, MD. et al.. (1989). Cells that overproduce protein kinase C are more susceptible to transformation by an activated H-ras oncogene. Mol. Cell Biol., 922641-2647. Huberman, E., and Sachs, L. (1974). Cell-mediated mutagenesis of mammalian cells with chemical carcinogens. Int. J. Cancer, 132326-333. Huberman, E., and Sachs, L. (1976). Mutability of different genetic loci in mammalian cells by metabolically activated carcinogenic polycyclic hydrocarbons. Proc. Natl. Acad. Sci. USA, 73:188-192. Huebner, R.J., and Todaro, G.J. (1969) Oncogenes of RNA tumor viruses as determinants of cancer. Proc. Natl. Acad. Sci. USA, 6421087-1094. Hunter, T., and Pines, J. (1994). Cyclins and cancer. ll: Cyclin D and CDK inhibitors come of age. Cell, 79: 573-582. Hunter, T. (1997). Oncoprotein networks. Cell, 882333-346. Hunter, T. (1991). Cooperations between oncogenes. Cell, 642249-270. Hunter, T. (1987). A thousand and one protein kinases. Cell, 502 823-829. HUpp, TR, and Lane, DP. (1994). Allosteric activation of latent p53 tetramers. Curr. Biol., 42865-875. lannaccone, P.M., Weinberg, WC, and Deamant, FD. (1987). On the clonal origin of tumors: a review of experimental models. Int. J. Cancer, 39:778-784. lchii, S., Takeda, S., Horii, A, Nakatsuru, S., Miyoshi, Y., Emi, M., Fujiwara, Y., Koyama, K., Furuyama, J., Utsunomiya, J., et al. (1993). Detailed analysis of genetic alterations in colorectal tumors from patients with and without familial adenomatous polyposis (FAP). Oncogene, 82 2399-405. Kallioniemi, A, Kallioniemi, O.P., Sudar, D., Rutovitz, D., Gray, J.W., Waldman, F., and Pinkel, D. (1992). Comparative genomic hybridization for molecular cytogenetic 66 analysis of solid tumors. Science, 258:818-821. Kallioniemi, OF. (1997). Linking chromosomal clues. Nature Genet, 1525-6. Kamb, A, Gruis, N.A, Weaver-Feldhaus, J., Liu, 0., Harshman, K., tavigian, S.V., Stochert, E., Day, R.S.Ill, Johnson, BE, and Skolnick, M.H. (1994). A cell cycle regulator potentially involved in genesis of many tumor types. Science, 264:436-440. Katsumata, M., Siegel, R.M., Louie, D.C. et al.. (1992). Differential effects of Bcl-2 on T and B cells in transgenic mice. Proc. Natl. Acad. Sci. USA, 89:11376-11380. Kawai, S., and Hanafusa, H. (1971). The effects of reciprocal changes in temperature on the transformed state of cells infected with a rous sarcoma virus mutant. Virology, 46: 470-479. Khazaie, K., Dull, T.J., Graf, T., Schlessinger, J., Ullrich, A, Beug, H., and Vennstomi, B. (1988). Truncation of the human EGF receptor leads to different transforming potentials in primary avian fibroblasts and erythroblasts. EMBO J., 723061-3071. Khosravi-Far, R., Solski, P.A, Clark, G.J., Kinch, MS, and Der, C.J. (1995). Activation of Rac1, RhoA, and mitogen-activated protein kinases is required for ras transformation. Mol. Cell Biol., 152 6443-6453. Khosravi-Far, R., White, M. A, Westwick, J.K., Solski, P.A, Chrzanowska-Wodnicka, M., Van-Aelst, L., Wigler, M.H., and Der, C.J. (1996). Oncogenic ras activation of Raf/mitogen-activated protein kinase-independent pathways is sufficient to cause tumorigenic transformation. Mol. Cell Biol., 16: 3923-3933. Kikkawa, U., Kishimoto, A, and Nishizuka, Y. (1989). The protein kinase C family: Heterogeneity and its implications. Annu. Rev. Biochem., 58:31-44. Kinzler, K.W., Nilbert, M.C., Su, L.K., Vogelstein, B., Bryan, T.M., Levy, D.B., Smith, K.J., Preislnger, AC., Hedge, P., McKechnie, D., et al. (1991). Identification of FAP locus genes from chromosome 5q21. Science, 253: 661-665. Kinzler, K.W., and Vogelstein, B. (1996). Life and death in a malignant tumor. Nature, 379219-20. Kitayama, H., Sugimoto, Y., Matsuzaki, T., lkawa, Y., and Noda, M. (1989). A ras- related gene with transformation suppressor activity. Cell, 56277-84. Klein, G. (1981). The role of gene dosage and genetic transposition in carcinogenesis. Nature, 294:313-318. Klein, G. (1987). The approaching era of the tumor suppressor genes. Science, 238:1539-1545. Klinger, HP. (1982). Suppression of tumorigenicity. Cytogenet. Cell Genet, 32:68-84. 67 Knudson, AG. (1971). Mutation and cancer: statistical study of retinoblastoma. Proc. NatI.Acad. Sci. USA. 68:820-823. Knudson, AG. (1986). Genetics of human cancer. Ann. Rev. Genet, 20:231-251. Knudson, AG. (1993). All in the cancer family. Nature Genet, 52103-104. Knudson, A. G. (1989). Hereditary cancers: Clues to mechanisms of carcinogenesis. Brit. J. Cancer, 59:661-666. Knudson, AG. Genetic predisposition to cancer, in Hiatt, H.H., Watson, JR, and Winsten, J.A(eds). Origin of Human Cancer, Cold Spring Harbor, NY: Cold Spring Harbor Laboratory, 1977, pp.45-52. Konopka, J.B., Watanabe, S.M., Witte, ON. (1984). An alteration of the human c-abl protein in K562 leukemia cells unmasks associated protein tyrosine kinase activity. Cell, 3721035. Kovary, K., and Bravo, R. (1991). The jun and fos protein families are both required for cell cycle progression in fibroblasts. Mol. Cell Biol., 11: 4466-4472. Krengel, U., Schlichting, L., Scherer, A, Schumann, R., Frech, M., John, J., Sabsch, W., Pai, BF, and Wittinghofer, A. (1990). Three-dimensional structures of H-ras p21 mutants: Molecular basis for their inability to function as signal switch molecule. Cell, 62:539-548. Kress, S., Sutter, C., Strickland, P.T., Mukhtar, H., Schweizer, J., and Schwartz, M. (1992). Carcinogen-specific mutational pattern in the p53 gene in ultraviolet B radiation- induced squamous cell carcinomas of mouse skin. Cancer Res., 52:6400-6403. Krontiris, T.G. and Cooper, GM. (1981). Transforming activity of human tumor DNAs. Proc. Natl. Acad. Sci. USA, 78:1181-1184. Krystal, G.W., Hines, S.J., and Organ, CF. (1996). Autocrine growth of small cell lung cancer mediated by coexpression of c-kit and stem cell factor. Cancer Res., 56:370- 376. Kuroki, T., and Huh, N-h. (1993). Why are human cells resistant to malignant cell transformation in vitro. Jpn. J. Cancer Res., 84:1091-1100. Lane, DP. (1994). p53 and human cancers. Brit. Med. Bulletin., 50:582-599. Lathangue, MB. (1994). DRTF1IE2F2 an expanding family of heterodimeric transcription factors implicated in cell-cycle control. Trends Biochem. Sci., 192108-114. Lawley, PD. (1989). Mutagens as carcinogens: development of current concepts. Mutation Res., 21323-25. Leder, P., Battey, J., Lenoir, G., Moulding, G, Murphy, W., Potter, H., Stewart, T., and 68 Taub, R. (1983). Translocation among antibody genes in human cancer, Science, 222:765-771. Lee, W-H., Boohstein, R., Hong, P., Young, L-J., Shew, J-Y., and Lee, E.Y-H.P. (1987). Human retinoblastoma susceptibility gene: cloning, identification, and sequence. Science, 235:1394-1399. Lee, S.W., Tomasetto, C., and Sager, R. (1991). Positive selection of candidate tumor suppressor genes by subtractive hybridization. Proc. Natl. Acad. Sci. USA, 8822825- 2829. Leonard, D.G.B., Ziff, EB, and Greene, L.A(1987). Identification and characterization of mRNAs regulated by nerve growth factors in PC12 cells. Mol. Cell Biol., 723156- 3167. Levine, AJ. (1993). The tumor suppressor genes. Annu. Rev. Biochem., 62:623-651. Levine, A.J. (1997). p53, the cellular gatekeeper for growth and division. Cell, 88:323- 331. Lewin, B. (1991). Oncogenic conversion by regulatory changes in transcription factors. Cell, 64:303-312. Li, PP (1988). Cancer families: Human models of susceptibility to neoplasia-the Richard and Hinda Rosenthal Foundation Award Lecture. Cancer Res., 4825381-5386. Liang, P., and Pardee, A. B. (1992). Differential display of eukaryotic messenger RNA by means of the polymerase chain reaction. Science, 257: 967-969. Liang, P., Averboukh, L., and Pardee, A. B. (1993). Distribution and cloning of eukaryotic mRNA by means of differential display: refinements and optimization. Nucleic Acids Res., 21 :3269-3275. Lin, C., Maher, V.M., and McCormick, J.J. (1995) Malignant transformation of human fibroblast strain MSU-1.1 by v-fes requires an additional genetic change. Int. J.Cancer., 63: 140-7 Lin, J., Wu, X., Chen, J., Chang, A, and Levine, A.J. (1995). Functions of the p53 protein in growth regulation and tumor suppression. Cold Spring Harbor Symposia on Quantitative Biology LIX, 215-223. Lindahl,T. (1994). DNA surveillance defect in cancer cells. Current Bi0109Y. 4:249. Lipponen, P., and Eskelinen, M. (1994). Expression of epidermal growth factor receptor in bladder cancer as related to established prognostic factors, oncoprotein (c-erbB-2, p53) expression and long-term prognosis. Brit. J. Cancer, 6921120-1125. Lovec, H., Grzeschiczek, A, Kowalski, MB, and Moroy, T. (1994). Cyclin D1/bcl-1 cooperates with myc genes in the generation of B-cell lymphoma in transgenic mice. 69 EMBO J., 13:3487-3495. Lowe, D.G., Capon, D.J., Delwart, E., Sakaguchi, A.Y., Naylor, S.L., and Goeddel, D.V. (1987). Structure of the human and murine R-ras genes, novel genes closely related to ras proto-oncogenes. Cell, 48: 137-146. Lowe, S.W., Schmitt, E.M., Smith, S.W., Osborne, BA, and Jacks, T. (1993). P53 is required for radiation induced apoptosis in mouse thymocytes. Nature, 362:847-849. Lu,H., and Levine, A.J. (1995). Human TAF-31 is a transcriptional coactivator of the p53 protein. Proc. Natl. Acad. Sci. USA, 92:5154-5158. Maher, V.M., Miller, E.C., Miller, J.A., and Szybalski, W. (1968). Mutations and decreases in density of transforming DNA produced by derivatives of the carcinogens 2-acetyl-aminofluorene and N-methyl-4-aminoazobenzene. Mol. Pharmacol., 42 411-426. Maki, Y., Bos, T.J., Davis, 0., Starbuck, M., and Vogt, PK. (1987). Avian sarcoma virus 17 carries the jun oncogene. Proc. Natl. Acad. Sci. USA, 84:2848-2852. Mandi, M., Paffenholz, R., Friedl, W., Caspari, R., Sengteller, M., and Propping, P. (1994). Frequency of common and novel inactivating APC mutations in 202 families with familial adenomatous polyposis. Hum. Mol. Genet, 3: 181-184. Margolis, B., Li, M, Koch, A, et al.. (1990). The tyrosine phosphorylated carboxyl terminus of the EGF receptor is a binding site for GAP and PLC-y. EMBO J., 924375- 4380. Markowitz, S., Wang, J., Myeroff, L., Parsons, R., Sun, L., Lutterbaugh, J., Fan, R. S., Zborowska, E., Kinzler, K.W., Vogelstein, B., et al.. (1995). Inactivation of the type II TGF-beta receptor in colon cancer cells with microsatellite instability. Science, 268: 1336-1338. Maron, D.M., and Ames, B.N. (1983). Revised methods for the Salmonella mutagenicity test. Mutat. Res., 1132173-215. Marshall, C.J. (1991). Tumor suppressor genes. Cell, 64:313-326. Marshall, MS. (1995). ras target protein in eukaryotic cells. FASEB J., 9:1311-1318. Martin, GS. (1970). Rous sarcoma virus: A function required for the maintenance of the transformed state. Nature, 22721021-1023. Maxwell, M., Naber, S.P., Wolfe, H.J., Galanopoulos, T., Hedley-Whyte, E.T., Black, P.M., and Antoniades, HM (1990). Coexpression of platelet-derived growth factor (PDGF) and PDGF-receptor genes by primary human astrocytomas may contribute to their development and maintenance. J. Clin. Invest, 86:131-140. McCann, J., Choi, E., Yamasaki, E., and Ames, B.N. (1975). Detections of carcinogens 70 as mutagens in the Salmonella/microsome test: Assay of 300 chemicals. Proc. Natl. Acad. Sci. USA, 7225135. McCormick, J.J., and Maher. V.M. (1994). Analysis of the multistep process of carcinogenesis using human fibroblasts. Risk Anal., 14:257-263. McCormick, F. (1993). How receptors turn ras on. Nature, 363215-16. McCormick, J.J., and Maher, V.M. (1988). Towards an understanding of the malignant transformation of diploid human fibroblasts. Mutation Res., 199:273-291. McDonnell, T.J., Troncoso, P., Brisbay, SM, et al.. (1992). Expression of the proto- oncogene bcI-2 in the prostate and its association with emergence of androgen- independent prostate cancer. Cancer Res., 52: 6940-6944. McDonnell, T.J., and Korsmeyer, SJ. (1991). Progression from lymphoid hyperplasia to high-grade malignant lymphoma in mice transgenic for the t(14;18). Nature, 3492254- 256. McGIade, J., Cheng, A, Pelicci, G., Pelicci, PG, and Pawson, T. (1992). She proteins are phosphorylated and regulated by the v-Src and v-Fps protein-tyrosine kinases. Proc. Natl. Acad. Sci. USA, 89:8869-8873. Miki, T., Fleming, T.P., Crescenzi, M., Molloy, C.J., Blam, S.B., Reynolds, SH, and Aaronson, SA (1991). Development of a highly efficient expression cDNA cloning system: application to oncogene isolation. Proc. Natl. Acad. Sci. USA, 882 5167-71. Miki, T., and Aaronson, SA (1995). Isolation of oncogenes by expression cDNA cloning. Methods Enzymol., 254: 196-206. Miller, EC. (1978). Some current perspectives on chemical carcinogenesis in human and experimental animals: Presidential Address. Cancer Res., 3821479. Miller, EC, and Miller, J.A. (1981). Searches for ultimate chemical carcinogens and their reaction with cellular macromolecules. Cancer, 4722327. Miyashita, T., and Reed, JC. (1993). Bcl-2 oncoprotein blocks chemotherapy-induced apoptosis in a human leukemia cell line. Blood, 812151-157. Modrich, P. (1994). Mismatch repair, genetic instability, and cancer. Science, 26621059- 1060. Moore, R., Casey, G., Brookes, 8., Dixon, M., Peters, G., and Dickson, C. (1986). Sequence, topography and protein coding potential of mouse int-2: a putative oncogene activated by mouse mammary tumor virus. EMBO J., 5: 919-24. Morgan, T.L., Yang, D.J., Fry, D.G., Hurlin, P.J., Kohler, S.K., Maher, V.M., McCormick, J.J. (1991). Characteristics of an infinite life span diploid human fibroblast cell strain and a near-diploid strain arising from a clone of cells expressing a transfected v-myc 71 oncogene. Exp. Cell Res.,197: 125-136. Morgan, DO. (1995). Principle of CDK regulation. Nature, 374:131-134. Motokura, T., Bloom, T., Kim, H.G., Juppner, H., Rudennan, J.V., Kronberg, HM, and Arnold, A. (1991). A novel cyclin encoded by a bcI-1 linked candidate oncogene. Nature, 3502512-515. Mottram, J. C. (1944). A developing factor in experimental blastogenesis. J. Pathol. Bacterial, 562181-187. Namba,M., Nishitani,K., Fukushima, F., Kimoto, T. (1988). Multistep carcinogenesis of normal human fibroblasts. Human fibroblasts immortalized by repeated treatment with Co-60 gamma rays were transformed into tumorigenic cells with Ha-ras oncogenes. Anticancer Res., 82 947-958 Nehls, M., Pfeifer, D., and Boehm, T. (1994). Exon amplification from complete libraries of genomic DNA using a novel phage vector with automatic plasmid excision facility: application to the mouse neurofibromatosis-1 locus. Oncogene, 92 2169-2175. Nelson, M.A, Futscher, B.W., Kinsella, T., Wymer, J., and Bowden, GT (1992). Detection of mutant Ha-ras genes in chemically initiated mouse skin epidermis before the development of benign tumors. Proc. Natl. Acad. Sci. USA, 89:6398-6402. Nevins, JR. (1992). E2F: a link between the Rb tumor suppressor protein and viral oncoproteins. Science, 2582424-429. Nicolaides, N.C., Papadopoulos, M, Liu, B., Wei, Y.F., Carter, K.C., Ruben, S.M., Rosen, C.A., Haseltine, W.A, Fleischmann, R.D., Fraser, C.M., et-al. (1994). Mutations of two PMS homologues in hereditary nonpolyposis colon cancer. Nature, 371: 75-80. Nishikawa, R., Ji, XD., Harmon, R.C., lazar, C.S., Gill, G.N., Cavenee, W.K., and Huang, H.J. (1994). Proc. Natl. Acad. Sci. USA, 9127727-7731. Nishizuka, Y. (1992). Intracellular signaling by the hydrolysis of phospholipids and activation of protein kinase C. Science, 2582607. Nobori, T., Miura, K., Wu, D.J., Lois, A, Takabayashi, K., and Carson, DA (1994). Deletion of the cyclin-dependent kinase 4 inhibitor gene in multiple human cancers. Nature, 368:753-756. Noda, M. (1990). Expression cloning of tumor suppressor genes: A guide for optimists. Moi. Carcinogenesis, 32251-253. Noda, M., Selinger, Z., Scolnick, EM, and Bassin, RH. (1983). Flat revertants isolated from kirsten sarcoma virus-transformed cells are resistant to the action specific oncogenes. Proc. Natl. Acad. Sci. USA, 8025602-5606. Okamoto, A, Demetrick, D.J., Spillare, E.A., Hagiwara, K., Hussein, S.P., Bennett, 72 WP, Forrester, K., Gerwin, B., Serrano, M., Beach, DH, and Harris, CC. (1994). Mutations and altered expression of p161NK4 in human cancer. Proc. Natl. Acad. Sci. USA, 91:11045-11049. O’Connell, J.F., Klein-Szanto, A.J.P., Digiovanni, D.M., Fries, J.W., and Slaga, T.J. (1986). Malignant progression of mouse skin papillomas treated with ethylnitrosourea, N-methyI-N’-nitro-N-nitrosoguanidine, or 12-O-tetradecanoylphorbol-13-acetate. Cancer Letter, 302269-274. Packham, G., and Cleveland, J.L. (1995). C-Myc and apoptosis. Biochim. Biophy. Acta, 1242:11-28. 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, MD, et al. (1994). Mutation of a mutL homolog in hereditary colon cancer. Science, 2632 1625-1629. Pardee, AB. (1989). G1 events and regulation of cell proliferation. Science, 2462603- 608. Pelicci, G., Lanfrancone, L., Grignani, F., McGIade, J., Cavallo, F., et al., (1992). A novel transforming protein (Shc) with an SH2 domain is implicated in mitogenic signal transduction. Cell, 70293-104. Perucho, M., Goldfarb, M., Shimizu, K., Lama, C., Fogh, J., and Wigler, M. (1981). Human-tumor-derived cell lines contain common and different transforming genes. Cell, 27:467-476. Peto, R. (1977). Epideology, multistage models, and short term mutagenesis tests. In Origins of Human Cancer. Cold Spring Harbor Laboratory, Cold Spring Harbor, NY,1403-1428. Pezzella, F., Turley, H., Kuzu, I., et al.. (1993). Bel-2 protein in non-small cell lung carcinoma. New Engl. J. Med., 329:690-694. Phuchareon, J., and Tokuhisa, T. (1995). Deregulated c-FosIAP-1 modulates expression of the cyclin and the cdk gene in splenic B cells stimulated with Iipopolysaccharide. Cancer Lett, 92: 203-208. Plowman, G.D., Green, J.M., Culouscou, J.M., Carlton, G. W., Rothwell, V.M., and Buckley, S. (1993). Heregulin induces tyrosine phosphorylation of HER4Ip180erbB4. Nature, 3662 473-475. Pollock, P.M., Pearson, JV, and Hayward, MK. (1996). Compilation of somatic mutations of the CDKN2 gene in human cancers: non-random distribution of base substitutions. Genes Chromosome Cancer, 15277-88. Powell, S.M., Petersen, G.M., Krush, A.J., Booker, S, Jen, J., Giardiello, F.M., Hamilton, S.R., Vogelstein, B., and Kinzler, K.W. (1992). Molecular diagnosis of familial adenomatous polyposis. N. Engl. J. Med., 329: 1982-987. 73 Qiu, R.G., Chen, J., Kim, D., McCormick, F., and Symons, M. (1995). An essential role for Rac in ras transformation. Nature, 3742 457-459. Quintanilla, M., Brown, K., Ramsden, M., and Balmain, A. (1986). Carcinogen-specific mutation and amplification of Ha-ras during mouse skin carcinogenesis. Nature, 322: 78-80. Rabbits, TH. (1994). Chromosomal translocations in human cancer. Nature, 372:143- 149. Rapp, U.R. (1990). Role of Raf-1 serine/threonine protein kinase in growth factor signal transduction. Oncogene, 62495-500. Reed, J.C., Cuddy, M., Slabiak, T., Croce, OM, and Nowell, PC. (1988). Oncogenic potential of bcl-2 demonstrated by gene transfer. Nature, 336: 259-61. Renan, M.J. (1993). How many mutations are required for tumorigenesis? Implications from human cancer data. Mol. Carcinogenesis, 72139-146. Rhim, J.S. (1993). Neoplastic transformation of human cells in vitro. Crit. Rev. Oncol., 42313-335. Riccardi, V.M., Hittner, H.M., Francke, U., et al.. (1990). The aniridia-Wilms’ tumor association: The critical role of chromosome band 11p13. Cancer Genet. Cytogenet., 22131-137. Rosenberg, C.L., Wong, E., Petty, E.M., Bale, AE., Tsujimoto, Y., Harris, ML, and Arnold, A. (1991b). PRAD1, a candidate BCL1 oncogene: mapping and expression in centrocytic lymphoma. Proc. Natl. Acad. Sci. USA, 88:9638-9642. Rosenberg, C.L., Kim, H.G., Shows, T.B., Kronberg, HM, and Arnold, A. (1991a). Rearrangement and overexpression of D118287E, a candidate oncogene on chromosome 11q13 in benign parathyroid tumors. Oncogene, 62449-453. Rotin, D., Margolis, B., Mohammadi, M., Daly, R.J., Daum, G., Li, M, Fischer, E. H., Burgess, W.H., Ullrich, A, and Schlessinger, J. (1992). SH2 domains prevent tyrosine dephosphorylation of the EGF receptor identification of Tyr992 as the high-affinity binding site for SH2 domains of phospholipase C gamma. EMBO J., 11:559-567. Rous, P. (1911) A sarcoma of the fowl transmissible by an agent separable from the tumor cells. J. Exp. Med, 13:397-411. Rous, P. (1910) A transmissible avian neoplasm: sarcoma of the common fowl. J. Exp. Med., 12:696. Ruddon, R.W. (1995). In Cancer Biology. Oxford University Press, pp.62-69. Ruggeri, B., Caamano, J., Goodrow, T., Dirado, M., Bianchi, A, Trono, D., Conti, C.J., and KIein-Szanto, A.J. (1991). Alterations of the p53 tumor suppressor gene during 74 mouse skin tumor progression. Cancer Res., 51 :6615-6621. Ruggeri, B., Dirado, M., Zhang, S.Y., Bauer, B., Goodrow, T., and Klein-Szanto, A.J. (1993). Benzo(a)pyrene-induced murine skin tumors exhibit frequent and characteristic G to T mutations in the p53 gene. Proc. Natl. Acad. Sci. USA, 9021013-1017. Ruther, U., Komitowski, D., Schubert, ER, and Wagner, E.F. (1989). C-fos expression induced bone tumors in transgenic mice. Oncogene, 42861-865. Ruther, U., Garber, C., Komitowski, D., Muller, R., and Wagner, E.F. (1987). Deregulated c-fos expression interferes with normal bone development in transgenic mice. Nature, 325:412-416. Saez, R., Chan, AM., Miki, T., and Aaronson, SA (1994). Oncogenic activation of human R-ras by point mutations analogous to those of prototype H-ras oncogenes. Oncogene, 92 2977-2982. Saez, E., Rutberg, S., Mueller, E., Oppenheim, H., Smoluk, J., Yuspa, H., and Spiegelman, BM. (1995). C-fos is required for malignant progression of skin tumors. Cell, 822721-732. Sager, R. (1985). Genetic suppression of tumor formation. Adv. Cancer Res., 44:43- 68. Sager, R. (1989). Tumor suppressor genes: The puzzle and promise. Science, 246:1406-1412. Sager, R. (1997). Expression genetics in cancer: Shifting the focus from DNA to RNA. Proc. Natl. Acad. Sci. USA, 94:952-955. Samuels, M.L., Weber, M. J., Bishop, J. M., and McMahon, M. (1993). Conditional transformation of cells and rapid activation of the mitogen-activated protein kinase cascade by an estradiol-dependent human raf-1 protein kinase. Mol. Cell Biol., 13: 6241-6252. Santos, E., Tronick, S.R., Aaronson, S.A, Pulciani, S., and Barbacid, M. (1982). T24 human bladder carcinoma oncogenes is an activated form of the normal human homologue of BALB and Harvey-MSV transforming genes. Nature, 2982343. Sargent, TD. (1987). Meth. Enzymol., 152:423-431. Sarma, V., Wolf, F.W., Marks, R.M., Shows, TB, and Dixit, V.M. (1992).Cloning of a novel tumor necrosis factor-alpha-inducible primary response gene that is differentially expressed in development and capillary tube-like formation in vitro. J lmmunol., 148: 3302-3312. Sassone-Corsi, P. (1992). Fatal attraction: The fos-jun affair with nuclear receptors. lntl. J.Oncol., 1:5. 75 Schaefer, R., lyer, J., lten, E., and Nirkko, A. (1988). Partial reversion of the transformed phenotype in HRAS-transfected tumorigenic cells by transfer of a human gene. Proc. Natl. Acad. Sci. USA, 85:1590-1594. Schlessinger, J., and Ullrich, A. (1992).Growth factor signaling by receptor tyrosine kinases. Neuron, 9: 383-91. Schmid, M., Haaf, T., and Grunert, D. (1984). 5-Azacytidine-induced undercondensations in human chromosomes. Hum. Genet, 67:257-263. Schuh, A.C., Keating, S.J., Monteclaro, F.S., Vogt, PK, and Breitman, ML. (1990). Obligatory wounding requirement for tumorigenesis in v-jun transgenic mice. Nature, 3462756-760. Schwab, M., Varmus, HE, and Bishop, J.M. (1985). Human N-myc gene contributes to neoplastic transformation of mammalian cells in culture. Nature, 316: 160-162. Seger, R., and Krebs, EC. (1995). The MAPK signaling cascade. FASEB J., 92726- 735. Sentman, C.L., Shutter, J.R., Hockenbery, D., Kanegawa, O., and Korsmeyer, SJ. (1991). Bcl-2 inhibits multiple forms of apoptosis but not negative selection in thymocytes. Cell, 67:879-888. Serrano, M., Lee, H-W., Chin, L., Cordon-Cardo, C., Beach, 0., and DePinho, RA (1996). Role of the INK4a locus in tumor suppression and cell mortality. Cell, 85:27-37. Seto, M., Yamamoto, K., lida, S., Akao, Y., Utsumi, K.R., Kubonishi, l., Miyoshi, l., Ohtsuki, T., yawata, Y., Namba, M., Motokura, T., Arnold, A, Takahashi, T. And Ueda, R. (1992). Gene rearrangement and overexpression of PRAD1 in lymphoid malignancy with t(1 1 ;14) (q13;q32) translocation. Oncogene, 721401-1406. Shay, J.W., Wright,W.E., Werbin, H. (1991). Defining the molecular mechanisms of human cell immortalization. Biochim. Biophys. Acta, 1072: 1-7. Shay, J.W., and Wright, W.E. (1989). Quantitation of the frequency of immortalization of normal human diploid fibroblasts by SV40 large T-antigen. Exp. Cell Res., 1842109. Sherr, C.J. (1990). Colony-stimulating factor-1 receptor. Blood, 75:1-12. Sherr, C.J. (1993). Mammalian G1 cyclins. Cell, 73:1059-1065. Sherr, C. J. (1994). G1 phase progression: cycling on cue. Cell, 79: 551-555. Sherr, C.J., and Roberts, J.M. (1995). Inhibitors of mammalian G1 cyclin-dependent kinases. Genes Dev., 921149-1163. Sherr, C.J. (1996). Cancer cell cycle. Science, 274:1672-1677. 76 Shih, C., Padhy, L.C., Murray, M., and Weiberg, RA (1981). Transforming genes of carcinomas and neuroblastomas introduced into mouse fibroblasts. Nature, 2902261- 264. Shih, C., Shilo, B-Z., Goldfarb, M.P., Dannenberg, A, and Weinberg, RA (1979). Passage of phenotypes of chemically transformed cells via transfection of DNA and chromatin. Proc. Natl. Acad. Sci. USA, 76:5714-5718. Shih, C., and Weiberg, RA (1982). Isolation of a transforming sequence from a human bladder carcinoma cell line. Cell, 29:161-169. Shirai, H., Ueno, E., Osaki, M., Tatebe, S., Ito, H., Kaibara, N. (1995). Expression of growth factors and their receptors in human early colorectal carcinomas: immunohistochemical Study. Anticancer Res., 15:2889-2894. Simon, H-G., Risse, B., Jost, M., Oppenheimer, 8., Karl, C., and Rodeck, U. (1996). Identification of differentially expressed messenger RNAs in human melanocytes and melanoma cells. Cancer Res., 56:3112-3117. Slamon, D.J., Godophin, W., Jones, L.A, Holt, J.A., Wong, S.G., Keith, D.E., Levin, W.J., Stuart, S.G., Udove, J., Ullrich, A, and Press, MP. (1989). Studies of the Her2/neu proto-oncogene in human breast and ovarian cancer. Science, 244:707-712. Smeyne, R.J., Vendrell, M., I-layward, M., et al., (1993). Continuous c-fos expression precedes programmed cell death in vivo. Nature, 363:166-169. Smith, JR, and Pereira-Smith, OM. (1996). Replicative senescence: implication for in vivo aging and tumor suppression. Science, 2732 63-67. Smith, M.J., and Prochownik, E. V. (1992). Inhibition of c-jun causes reversible proliferative arrest and withdrawal from the cell cycle. Blood, 79: 2107-2115. Sobel, ME. (1990). Metastasis suppressor genes. J. Natl. Cancer Inst, 82: 267-276. Solomon. E., Borrow, J., Goddard, AD. (1991). Chromosome aberrations and cancer. Science, 25421 153-1 160. Spector, D.H., Varmus, HE, and Bishop, J.M. (1978a). Nucleotide sequences related to the transforming gene of avian sarcoma virus are present in DNA of uninfected vertebrates. Proc. Natl. Acad. Sci.USA, 75: 4102-4106. Spector, D.H., Smith, K., Padgett, T., McCombe, P., Roulland-Dussoix, D., Moscovic, C., Varmus, HE, and Bishop, J.M. (1978b). Uninfected avian cells contain RNA related to the transforming gene of avian sarcoma viruses. Cell, 13:371-379. Sporn, MB, and Todaro, G.J. (1980). Autocrine secretion and malignant transformation of cells. N. Engl. J. Med., 303:878-880. Srivastava, S., Zou, Z., Pirollo, K., Blattner, W., and Chang, EH. (1990). Gemi-line 77 transmission of a mutated p53 gene in a cancer-prone family with Li-Fraumeni syndrome. Nature, 348:747-749. Stanbridge, E.J., Flanderrneyer, R.R., Daniels, 0., and Nelson-Rees, WA (1981). Specific chromosome loss associated with the expression of tumorigenicity in human cell hybrids. Somatic Cell Genet, 7:699-712. Stehelin, D., Guntaka, R.V., Varmus, HE, and Bishop, J.M. (1976a). Purification of DNA complementary to nucleotide sequences required for neoplastic transformation of fibroblasts by avian sarcoma viruses. J. Mol. Biol., 1012 349-365. Stehelin, D., Varmus, H., Bishop, J.M., and Vogt, P.K. (1976b) DNA related to the transforming genes of avian sarcoma viruses is present in normal avian DNA. Nature, 2602170-173. Stephens, R.M., Sithanandam, G., Copeland, T.D., Kaplan, D.R., Rapp. U.R., and Morrison, D.K. (1992). 95-kilodalton B-Raf serine/threonine kinase: identification of the protein and its major autophosphorylation site. Mol. Cell Biol., 12: 3733-3742. Storm, SM, and Rapp, U.R. (1993). Oncogene activation: c-raf-1 gene mutations in experimental and naturally occurring tumors. Toxicol. Lett, 67: 201-210. Storm, S.M., Brennscheidt, U., Sithanandam, G., and Rapp, U.R. (1990). raf oncogenes in carcinogenesis. Crit. Rev. Oncog., 2: 1-8. Strasser, A, Harris, A.W., Bath, M.L., and Cory, S. (1990). Novel primitive lymphoid tumors induced in transgenic mice by cooperation between myc and bcl-2. Nature, 3482331-333. Strasser, A, Harris, A.W., and Cory, S. (1991). Bcl-2 transgene inhibits T cell death and perturbs thymic self-censorship. Cell, 67:889-899. Sun, Y., Hegamyer, G., and Colbum, N. H.(1994). Molecular cloning of five messenger RNAs differentially expressed in preneoplastic or neoplastic JB6 mouse epidermal cells: One is homologous to human tissue inhibitor of metalloproteinases-3. Cancer Res., 5421 139-1 144. Swenson, D.H., Petzold, G.L., and Harbach, PR. (1986). The binding of 1-(2-hydroxyethyl)-1-nitrosourea to DNA in vitro and to DNA of thymus and marrow in C57BL mice in vivo. Cancer Lett, 33: 75-81. Taub, R., Moulding, C., Battey, J., Murphy, W., Vasicek, T., Lenoir, GM, and Leder, P. (1984). Activation and somatic mutation of the translocated c-myc gene in Burkitt lymphoma cells. Cell, 362339-348. Taub, R., Kirsch, l., Monton, C., Lenoir, 6., Swan, 0., Tronick, S., Aaronson, S., and Leder, P. (1982). Translocation of the c-myc gene into the immunoglobulin heavy chain locus in human Burkitt’s lymphoma and murine plasmacytoma cells. Proc. Natl. Acad. Sci. USA, 79:7837-7841. 78 Tong, L. A, DeVos, AM., Milbum, M.V., Jancarik, J., Noguchi, S., Nishimora, S., Miura, K., Ohtsuka, E., and Kim, S-H. (1989). Structural differences between a ras oncogene protein and the normal protein. Nature, 337290-93. Tooze, J. (Ed): DNA tumor viruses. Cold Spring Harbor, NY, Cold Spring Harbor Laboratory Press, 1980. Toyoshima, K., Friis, R. R., and Vogt, P. K. (1970). The reproductive and cell-transforming capacities of avian sarcoma virus B77: inactivation with UV light. Virology, 42: 163-170. Treisman, R.H., Novak, U., Favaloro, J., and Kamen, R. (1981) Transformation of rat cells by an altered polyoma virus genome expressing only the middle T protein. Nature, 292:595-600. Treisman, R. (1994). Ternary complex factors: growth factor regulated transcriptional activators. Curr. Opin. Genet. Dev., 4: 96-101 Tsujimoto, Y. Finger, L.R., Yunis, J., Nowell, PC, and Croce, CM. (1984). Cloning of the chromosome breakpoint of neoplastic B cells with the t(14;18) chromosome translocation. Science, 226:1097-1099. Ullrich, A, and Schlessinger, J. (1990). Signal transduction by receptors with tyrosine kinase activity. Cell, 61:203-212. Varmus, H. (1989). An historical overview of oncogene. In Oncogenes and Molecular Origins of Cancer. R. A. Weinberg, ed. (New York: Cold Spring Harbor Laboratory Press). pp.3-44. Varmus, HE. (1984). The molecular genetics of cellular oncogenes. Annual Rev. Genet, 18:553-612. Vaux, D.L., Cory, S., and Adams, J.M. (1988). Bel-2 gene promotes haemopoietic cell survival and cooperates with c-myc to immortalize pre-B cells. Nature, 335:440-442. Veale, D., Ashcroft, T., Marsh, 0., Gibson, G.J., and Harris, AL. (1987). Epidermal growth factor receptors in non-small cell lung cancer. Brit. J. Cancer, 55:513-516. Vega, Q.C., Cochet, C.C., Filhol, 0., Chang, C-P., Rhee, S.G., and Gill, G.N. (1992). A site of tyrosine phosphorylation in the C terminus of the epidermal growth factor receptor is required to activate phospholipase C. Mol. Cell Biol., 122128-135. Vogelstein, B., Fearon, E.R., Hamilton, 8., Kern, E., Preislnger, AC., Leppert, M., Nakamura, Y., White, R., Smits, AM.M., and Bos, J.L. (1988). Genetic alterations during colorectal tumor development. N. Engl. J. Med., 319:525-532. Vogt, PK (1994). Oncogenic transformation by Jun. In The Fos and Jun families of transcription factors. Angel, PE, and Hrrrlich, P.A, Eds., (CRC Press, Inc., Florida), pp.203-219. 79 Vojta, P.J., and Barrett, JC. (1995). Genetic analysis of cellular senescence. Biochim. Biophy. Acta, 1242229-41. Volm, M., Efferth, T., Mattem, J., and Wodrich, W. (1992). Overexpression of c-fos and c-erbB-1 encoded proteins in squamous cell carcinomas of the lung of smokers. Int. J. Oncol., 1269-71. Waga, S., Hannon, G.J., Beach, 0., and Stillman, B. (1994). The p21 cyclin-dependent kinase inhibitor directly controls DNA replication via interaction with PCNA. Nature, 369:574-578. Wagner, A.J., Kokontis, J.M., and Hey, N. (1994). Myc-mediated apoptosis requires wild type p53 in a manner independent of cell cycle arrest and the abilit6y of p53 to induce p21‘“"’°'°‘- Genes Dev., 822817-2830. Walker, RA, and Cowl, J. (1991). The expression of c-fos protein in human breast. J. Pathol., 1632323-327. Wang, T.C., Cardiff, R.D., Zukerberg, L., Lees, E., Arnold, A, and Schmidt, E.V. (1994). Mammary hyperplasia and carcinoma in MMTV-cyclin D1 transgenic mice. Nature, 369:669-671. Wang, Z.Q., Grigoriadis, AE., Mohle-Steinlein, U., and Wagner, E.F. (1991). A novel target cell for c-fos-induced oncogenesis: development of chondrogenic tumors in embryonic stem cell chimeras. EMBO. J., 10: 2437-2450. Waterfield, M.D., Scrace, G.T., Whittle, N., Stoobant, P., Johnson, A, Wasteson, A, Westermark, B., Heldin, C-H., Huang, J.S., and Deuel, TE (1983) Platelet-derived growth factor is structurally related to the putative transforming protein p28sis of simian sarcoma virus. Nature, 304235-39. Weber, J.L. (1990). Human DNA polymorphisms based on length variations in simple- sequence tandem repeats. Genome Anal., 12159-181. Weinberg, RA (1995) The retinoblastoma protein and cell cycle control. Cell, 81:323- 330. Weiner, D.B., Liu, J., Cohen, J.A., Vifilliams, W.V., and Greene, M. (1989). A point mutation in the neu oncogene mimics ligand induction of receptor aggregation. Nature, 3392230231. Weiss, W., Teich, N., Varmus, H., and Coffin, J. (Eds): RNA tumor viruses. Cold Spring Harbor, NY, Cold Spring Harbor Laboratory Press, 1982. Wicking, C., and Williams, B. (1991). From linked marker to gene. Trends Genet, 72288-293. Wodrich, W., and Volm, M. (1993). Overexpression of oncoproteins in non-small cell lung carcinomas of smokers. Carcinogenesis, 14:1121-1124. 80 Woolford, J.W., McAuliffe, A. and Rohrschneider, LR. (1988). Activation of the feline c- fms proto-oncogene: multiple alterations are required to generate a fully transformed phenotype. Cell, 55:965-977. Wright, W.E., and Shay, J.W. (1992). The two-stage mechanism controlling cellular senescence and immortalization. Exp. Gerontol., 27:383-389. Wu, X., and Levine, A.J. (1994). p53 and E2F-1 cooperate to mediate apoptosis. Proc. Natl. Acad. Sci. USA, 91:3602-3606. Xiong, Y., Menninger, J., Beach, D. And Ward, DC. (1992). Molecular cloning and chromosomal mapping of CCND genes encoding human D-type cyclins. Genomics, 13:575-584. Xiong, Y., Hannon, G.J., Zhang, H., Casso, D., Kobayashi, R., and Beach, D. (1993). p21 is a universal inhibitor of cyclin kinases. Nature, 366:701-704. Young, D., Waitches, G., Birchmeier, C., Fasano, O., and \Mgler, M. (1986). Isolation and characterization of a new cellular oncogene encoding a protein with multiple potential transmembrane domains. Cell, 452 711-719. Yaraden, Y., and Ullrich, A. (1988). Growth factor receptor tyrosine kinase. Annu. Rev. Biochem., 57:443-478. Yates, J.W., and Connor, J.M. (1986). Genetic Linkage. Brit. J. Hosp. Med, 133-136. Yunis, J.J., and Ransay, N. (1978). Retinoblastoma and subband deletion of chromosome 13. Am. J. Dis. Child, 132:161-163. Yunis, J.J., Oken, N., Kaplan, M.E., Ensrud, K.M., Howe, K.R., and Theoligides, A. (1982). Distinctive chromosomal abnormalities in histologic subtypes of non-Hodgkin’s lymphomas. N. Engl. J. Med, 307:1231-1236. Yuspa, SH, and Dlugosz, A A (1991). Cutaneous carcinogenesis: natural and experimental. In: LA. Goldsmith (ed), Physiology biochemistry and molecular biology of the skin., pp. 1365-1402. New York: Oxford University Press. Yuspa, SH. (1994). The pathogenesis of squamous cell cancer: Lessons learned from studies of skin carcinogenesis-Thirty-third G.H.A Clowes memorial award lecture. Cancer Res., 54:1178-1189. Zur Hausen, H. (1991). Viruses in human cancer. Science, 254:1167-1173. CHAPTER II Suppression of Anchorage-independent Growth and Matrigel Invasion and Delayed Tumor Formation by Elevated Expression of Fibulin-1D in Human Fibrosarcoma-derived Cell Lines Jing Qing, Veronica M. Maher, Huan Tran, W. Scott Argraves, Robert W. Dunstan, J. Justin McCormick Carcinogenesis Laboratory-Fee Hall Department of Biochemistry and Department of Microbiology The Cancer Center Michigan State University, East Lansing, MI 48824 81 82 ABSTRACT Using differential display, we identified an mRNA that is markedly down-regulated in cell line 6AISB1, derived from a fibrosarcoma formed in an athymic mouse following injection of carcinogen-transformed MSU-1.1 cells. The nontumorigenic parental cell strain, MSU-1.1, expresses high levels of this mRNA. Sequencing of the corresponding cDNA fragment revealed that it corresponded to an expressed sequence tag, which ultimately led to its identification as the fibula-JD gene. Fibulin-1 is a cysteine-rich, calcium-binding extracellular matrix and plasma protein, which has four isoforms A-D derived from alternative splicing. Northern and Western blotting analysis of 16 cell lines established from tumors formed in athymic mice by MSU-1.1-derived cell strains independently transformed in culture showed that 44% exhibited low level or lack of expression of fibulin-1D mRNA and protein. In a similar analysis of 15 malignant cell lines derived from patients, 80% showed low level or no expression. To study the role of fibulin-1D in transformation, we transfected 6AISB1 cells and a human fibrosarcoma- derived cell line (SHAC) with a fibulin-1D cDNA expression construct. Transfectants displaying high levels of fibulin-1D were isolated and characterized. Elevated expression of fibulin-1D led to reduced ability to form colonies in soft agar and reduced invasive potential as tested in a matrigel in vitro invasion assay. Furthermore, expression of fibulin-1D resulted in a markedly extended latency in tumor formation in athymic mice. These results indicate that low expression of fibulin-1D plays a role in tumor formation and invasion. 83 INTRODUCTION Extensive research over the past two decades has established that cancer in humans arises from a cell that has acquired multiple genetic alterations in oncogenes and/or tumor suppressor genes (Weinberg, 1989; Fearon and Vogelstein, 1990; Hunter, 1991). Mutations and/or altered expression of these critical genes and their downstream effector genes allow a cell to escape from normal growth control and become malignant. A direct comparison of gene expression between non-tumorigenic and tumorigenic cells of the same genetic background should provide information on genetic changes involved in the transformation process. Subsequent cloning and functional analysis of these differentially expressed genes may enrich our understanding of cancer development. To study the molecular mechanisms underlying neoplastic transformation of human cells, we used the near-diploid, karyotypically stable, infinite life span human fibroblast cell strain MSU-1.1 as a model system. Cell strain MSU-1.1 was established in this laboratory from a normal diploid human neonatal foreskin fibroblast cell line (Morgan et al., 1991). The cells are phenotypically normal and do not form tumors in athymic mice. Transfection of MSU-1.1 cells with an activated ras oncogene expressed at high levels (Hurlin et al., 1989; Vifllson et al., 1990) or treatment of the cells with chemical carcinogens, such as (:)-7[3,8a-dihydroxy-9a,10a-epoxy-7,8,9,10- tetrahydrobenzo[a]pyrene (BPDE) followed by selection for focus-forming cells (Yang at al., 1992) results in transformants capable of tumor formation in athymic mice. However, the molecular basis of the neoplastic conversion in MSU-1.1 cells by treatment with chemical carcinogens is not understood. To identify putative oncogene(s) and tumor suppressor gene(s), as well as other types of genes associated with malignant transformation, we used differential mRNA display (Liang and Pardee, 84 1992; Liang at al., 1993) to compare the profile of gene expression in parental cell strain MSU-1.1 and cell line 6AISB1. The latter was derived from a fibrosarcoma formed in an athymic mouse by a BPDE-transformed MSU-1.1 cell strain. One of the differentially expressed genes identified corresponds to fibulin-1 D, an extracellular matrix (ECM) and plasma protein (Argraves et al., 1990, Tran e1 al., 1997). It is expressed at high levels in MSU-1.1 cells and is markedly down-regulated in 6AISB1 cells. Analysis of 16 cell lines established from tumors formed in athymic mice by MSU-1.1-derived cell strains transformed in culture by various agents showed that seven (44%) had a low level or complete lack of expression of fibulin-1D mRNA and protein. Similarly, analysis of 15 cell lines derived from tumors taken from patients showed that 12 (80%) had a low or complete lack of expression of this mRNA and protein. To determine if low expression of fibulin-1D were causally involved in the malignant transformation of these cell lines, we studied the effect of stably expressing fibulin-1 D in human fibrosarcoma-derived cell line SHAC and in 6AISB1ceIIs, which lack the expression of this protein. The results showed that elevated expression of fibulin-1D significantly decreased the anchorage-independent growth of both SHAC and 6AISB1 cell derivatives, as well as their invasiveness when tested in a matrigel in vitro invasion assay, and significantly delayed the onset of tumor formation in athymic mice by these cell lines. 85 RESULTS To identify transformation-related genes, we carried out differential mRNA display analysis, comparing the non-tumorigenic parental MSU-1.1 cell strain and a tumorigenic derivative cell line, 6AISB1. RT-PCR reactions were carried out using 80 different combinations of primer sets, composed of four degenerate anchored oligo(dT) primers, T12MN (M was dG,dC or dA; N was dG, dC, dA or dT) and 20 arbitrary 10-mers (OPA 1- 20, Operon Technologies, Alameda, CA). About 8,000 bands were observed. The band pattern for cell line 6AISB1 differed from that of parental MSU-1.1 in nine bands, each of which was down-regulated in 6AISB1 cells. These cDNAs were excised from the sequencing gel, reamplified by PCR, and used as probes for Northern blot analysis. Such analysis showed that only five of these nine differentially-displayed cDNAs represented mRNAs that actually are expressed at significantly lower levels in the 6AISB1 cells as compared with the MSU-1.1 cells. One of the five unique bands, designated SG7, is characterized in this report. As shown in Figure 1B, this cDNA hybridized to a 2.7-kb transcript showing a 14-fold lower level of expression in 6AISB1 cells than in MSU-1.1 cells. Cloning and sequencing of this partial cDNA indicated that it consisted of 311 base pairs and was 98.7% identical to an expressed sequence tag (Genbank accession No. T19384). To get the full-length cDNA corresponding to SG7, EST clone T19384 was obtained from the investigator who reported it, Dr. Georges Guellaen (Hopital Henri Mondor, Creteil, France), and the 5' and the 3' regions of the EST clone were sequenced. A database search revealed that the EST corresponded to the human fibula-10 gene, with 97.4% identity in a 312-bp overlap in the 5' region (bases 1-312 of fibulin-1 D) and 98.9% identity in a 188-bp overlap in the 3' region (bases 2172-2359 of fibulin-1D). The 86 SG7 cDNA that we isolated by differential display contained a sequence that extended beyond the 3' sequence of the fibulin-1 D cDNA sequence deposited in the database. To confirm the identity of SG7 as human flbulin-1D, we carried out Northern blot analysis using the 2.3-kb human fibulin-1D cDNA insert from plasmid pBluescriptSKfibulin1 D (Tran et al., 1997) to probe the same membrane previously used to detect SG7 expression. As shown in Figure 10, the fibulin-1 D cDNA probe detected the 2.7-kb mRNA transcript that had been detected by SG7 (Fig.1B), but it also detected a 2.3-kb mRNA transcript which corresponds to fibulin-1 C. This is consistent with the fact that these two mRNAs have identical 5' regions (bases 1-1707). Both transcripts exhibited coordinately decreased expression in 6AISB1 cells compared with the parental MSU-1.1 cells. A. _ .H .4.. .. pf.- ' . “h . _ ”h ,... .- .I_ To test whether the decreased expression of fibulin-1 D transcript is a common feature of human tumor cells, we carried out Northern analysis using fibulin-1D cDNA as a probe with RNA from 15 cell lines derived from malignant tumors taken from patients and 16 cell lines established from tumors produced in athymic mice by MSU-1.1-derived cell strains transformed in culture by oncogene transfection or carcinogen treatment. The origin of these cell lines is listed in Table 1. Examples of results are shown in Figure 2. Very low levels of expression of fibulin-1 were found in five out of five fibrosarcoma-derived cell lines (lanes 15-19), in two out of two osteosarcoma-derived cells (lanes 8 and 9), in two out of three neurofibrosarcoma-derived cells (lanes 12 and 20), in one out of two human bladder carcinoma-derived cells (lane 11), and in the rhabdomyosarcoma- and cervical carcinoma-derived cell lines (lanes 7 and 10) tested in this study. Downregulation of fibulin-1 mRNA expression was also found in 6 out of 16 MSU-1.1 lineage-derived tumor cells (lanes 3-6, and 22-23), in addition to cell strain 87 6A/SBl, in which the downregulation was first identified (Fig. 1). Taken together, 19 out of 31 tumor-derived cell lines exhibited low or no expression of fibulin-1 (C and D transcripts). To determine whether mRNA levels correlate with fibulin-1 protein expression, we performed Western blot analysis of cell extracts and their serum-free conditioned culture media. As a control, purified fibulin-1 protein from human placenta was included in each assay. As shown in Figure 3, lane 1, under non-reducing conditions, mouse-anti- human fibulin-1 monoclonal antibody 3A11 specifically recognized the fibulin-1 protein (apparent molecular weight, 80 kDa). We found that, consistent with results from Northern analysis, the expression of fibulin-1 protein was very low or undetectable in a panel of tumor cell lines, not only in the cell extracts (Fig.3A) but also in their conditioned culture media (Fig.3B), whereas a neonatal foreskin-derived human fibroblast line, LG1, and MSU-1.1 cells expressed high levels of fibulin-1 proteins. The presence of multiple immunoreactive polypeptides that migrated faster than the mature fibulin-1 polypeptide on SOS-PAGE (Fig.3A) may correspond to proteolytic fragments of fibulin-1. Similar fragments have been observed in purified fibulin-1 preparations derived from placenta and lung and have been generated in vitro by matrilysin and leucocyte elastase digestion (Sasaki et a], 1996). ._.'...-. . .; .,.._.A : ..- ._. -,.- H...” - - . .._'.e DLQIQ'IIJ The levels of flbulin-1 protein in either cell extracts or conditioned media (Fig.3) from fibrosarcoma-derived cell lines SHAC and 6AISB1 is almost undetectable. To examine the effect of fibulin-1D expression on malignant transformation, we constructed a human fibulin-1D cDNA expression vector, designated pPuro-Fibulin-1D, and transfected it into SHAC and 6AISB1 cells. Puromycin resistant clones were isolated from cells that were 88 Fig.1. (A) Differential display comparing mRNAs from the parental MSU-1.1 cell strain (lanes 1 and 2) and its malignant derivative cell line 6AISB1 (lane 3 and 4). For each cell line, two independent preparations of total RNA were extracted, reverse transcribed, and amplified by PCR in the presence of [oz-35$]dATP. The PCR products were separated on a 6% polyacrylamide gel and autoradiographed. The signal demonstrating altered expression is marked by an arrow. Primers used were T12MG and OPA7. (B) Northern blot analysis confirming differential gene expression for fragment SG7 using the cloned cDNA fragment as a probe. Total RNA (15 pg) obtained from MSU-1.1 cells (lane 1) and 6AISB1 cells (lane 2) was subjected to Northern analysis as detailed in “Materials and Methods”. The blot was stripped and reprobed with a GAPDH cDNA (lower panel) as the loading control. (C) Northern blot analysis using fibulin-1 D cDNA as a probe. The same blot of (B) was stripped and hybridized with a radiolabeled 2.3-kb human fibulin-ID cDNA insert from the pBluescriptSKfibulin-1D plasmid. 89 + 4. . _ .2 2 mi; 3 - _.._ . 23.no- Boga 1.;3 8.; A 2 u . Fig.1 90 the result of independent transfection events, i.e., from independent dishes. lmmunoblot analysis of whole cell extracts was used to identify clones that expressed high levels of fibulin-1D protein. For SHAC cells, a total of 24 clonally-derived, drug- resistant populations were tested. Five independent clones exhibited high levels of fibulin-1D protein. Two clones of SHAC cells that had been transfected with the control vector pBABE-Puro and displayed no detectable fibulin-1D protein were used as negative controls. The same procedure was carried out with the 6AISB1 cells. Of 45 puromycin-resistant clones assayed, 10 showed high expression of fibulin-1D. Six independent positive clones and three vector-transfected negative control clones were selected at random for further characterization. We also evaluated the level of flbulin-1 secreted into the conditioned culture medium by these cell strains. As expected, neither the parental cells nor the vector-transfected control clones secreted fibulin-1. In contrast, the transfected clones that expressed high levels of cellular fibulin-1D secreted relatively high levels of mature proteins into the medium (Fig.4). ;. ...,._--'.._-.-..,-. eeule .- ..- .- -,.- pf..- Since most malignantly transformed cells, including SHAC and 6AISB1 cells exhibit anchorage-independent growth, i.e., do not require attachment to a substratum for proliferation, whereas normal human fibroblasts and MSU-1.1 cells do, we tested whether fibulin-1D expression affects the ability of SHAC and 6AISB1 cells to form colonies in agarose. As shown in Figure 5 and Table 2, all the transfectant clones expressing high levels of fibulin-1D exhibited a reduced ability to grow in agarose compared to their parental cells and their vector-transfected control cell strains. The SHAC cells were counted after 21 days; the 6AISB1 cells after 14 days. A substantial decrease in the size of colonies was observed for SHAC cell transfectants. A moderate 91 Table 1 Human tumor-derived cell lines tested for fibulin-1 expression Cell lines3 Origin of the cell lines L451lB5T, L46l/7T, L551l3T, 2FTfl'1 ZC1IST1, 6AISB1, 11C/SB1 MSU-1.1-y1- 2A1/T, MSU-1.1- y4-2A/SF1" MSU-1.2-y1/SF1, msu-1.2-yz/sr1 2MT, 3MT, 178-2DT, DW5T,1.1-sisB/ST2 wsu1rr1, WSU17/T1 NCI, VIPzFT, SHAC, HT1080, 8387° 143 BTK, TE85 RD Hela GM03808“ T24, A1698“ NF1.90.8/FT1' Derived from spontaneously transformed MSU-1.1cells Derived from BPDE-transformed MSU-1.1 cells Derived from 6°Co-transformed MSU-1.1 cells Derived from ”Co-transformed MSU-1.2 cells Derived from oncogene-transfonned MSU-1.1 cells Derived from human neurofibrosarcomas Derived from human fibrosarcomas Derived from human osteosarcomas Derived from a human rhabdomyosarcoma Derived from a human cervical carcinoma Derived from a Wilms' tumor Derived from human bladder carcinomas Derived from a human neurofibrosarcoma Cell lines in the first five lines of the table were established in this laboratory from sarcomas generated in athymic mice by injection of transformed MSU-1.1 cells; those on line six were established in this laboratory from human neurofibrosarcomas. The rest, with the exception of 8387, GM03803, A1698, and NF1.90.8/FT1, were obtained from American Type Culture Collection, Rockville, MD. Referred to as MSU-1.1-y1 in Figure 2. ‘OQOU' From Dr. Stuart A. Aaronson, National Cancer Institute, Bethesda, MD. From Coriell Institute for Medical Research, Camden, NJ. From Dr. 0. M. Pereira-Smith, Baylor College of Medicine, Houston, TX. From Dr. Thomas Glover, University of Michigan, Ann Arbor, MI. 92 Fig. 2. Northern blot analysis of fibulin-I expression in multiple human tumor cell lines. Total RNA (15 pg), obtained from a series of cell lines, was hybridized with fibulin-1D cDNA as in Figure 1C. These were: fibroblast cell lines from normal donors (lanes 1 and 2); malignant cell lines derived from tumors formed in athymic mice after injection of MSU-1.1 cells transformed by a transfected oncogene (lanes 3-5) or spontaneously transformed (lane 6); malignant cell line derived from a patient’s rhabdosarcoma (lane 7), osteosarcoma (lanes 8 and 9), cervical carcinoma (lane 10), bladder carcinoma (lane 11), neurofibrosarcoma (lane 12); the normal cell line from which MSU-1.1 cells are derived (lane 13); parental MSU-1.1 cells (lane 14); malignant cell lines derived from a patient's fibrosarcoma (lanes 15-19), or neurofibrosarcoma (lane 20); and malignant cell lines derived from athymic mice tumors formed after injection of cells transformed by cobalt 60, viz., MSU-1.1 cells (lane 21), MSU-1.2 cells (lanes 22 and 23). The KD1 cell line was originally obtained from the late Dr. Takeo Kakunaga. The other cell lines are described in Table 1. (B) RNA loading evaluated by a GAPDH cDNA probe. «or. mNNNPNONGmehwoFmFQFMF NFFFOF m m h o m w m NF gifieeommeJ-Jmcte-eeo-eno- 9 Essen/i I I s. .-. Dr .. : .- r e.! i _ Bessel H A W1Mi. I18 us EX swswummmmsmSflwmflammmmmm .nnmflnwzomi... .n m 19 Mm... till .I M 1 seems .. m. ._ uuui 94 reduction in the size of colonies was seen for 6AISB1 cell transfectants. These results indicate that elevated expression of fibulin-1D can partially suppress anchorage independence in the flbrosarcoma-derived cell lines tested. To analyze the invasive potential of the fibulin-1D transfectants, we used a modified Boyden Chamber Matrigel assay. As shown in Figure 6, the ability of the fibulin-1D expressing transfectants to reach the bottom of the filters through the matrigel was reduced by 50-60% compared to that of parental cells and the vector-transfected control cell strains of both SHAC and 6AISB1 cells. I . | ...| [llllll'-ID- . I To determine whether restoration of the expression of the fibulin-1D gene would also suppress in vivo tumor growth, we inoculated the fibulin-1D-expressing transfectant cell strains, their nontransfected parental cell lines, and vector-transfected controls into athymic mice and monitored the animals for growth of tumors. As shown in Figure 7, the latency of tumor formation for the fibulin-1D-expressing clones was longer than that of the parental cells and the vector-transfected control cell strains. For SHAC cells and their vector-transfected control cell strains, the tumor volume reached 500 mm3 after an average of 25 days postinjection; whereas for fibulin-1D-transfected cells, the average time was 55 days. For 6AISB1 cells and their vector-transfected control strains, with the exception of one, the tumor volume reached 500 mm3 after an average of 20 days postinjection; whereas the average time for four fibulin-1D-expressing clones was 48 days, and the other two fibulin-1D-expressing clones failed to form tumors in three months. i 'I I'll |' [HI l'-Il'.2| IE I l 95 Fig. 3. Western blot analysis of the expression and secretion of fibulin-1 by normal cells (LG1 and MSU-1.1) and 10 tumor-derived cell lines under non-reducing conditions. Cellular protein (20 pg) (A) and 50 pl of the medium conditioned by the same cell lines (B) were analyzed by electrophoresis in 10% polyacrylamide gel and probed with a monoclonal antihuman fibulin-1 antibody 3A11. The positive control (lane 1) was fibulin— 1 protein isolated and purified from human placenta; lanes 4-8 are from malignant cell lines derived from human fibrosarcomas from patients; lane 9 from a rhabdomyosarcoma; lane 10 from an osteosarcoma; lanes 11-13 are from malignant cell lines derived from tumors in athymic mice from carcinogen-transformed MSU-1.1 cell strains. 96 —80 KD I-:ISII»l 'Z'I 1W8 I-BSN9 9831. G H 080 LLH L888 OVI-IS szlA ION l'I-‘nSW I91 I- uII"‘I!:I - “'80 KD Fig.3 97 If the transfected cells expressing fibulin-1 D grew more slowly than the parental cells, this could explain the biological data described above. To determine the doubling time of each of the fibrosarcoma—derived cell lines, the parental cells, two vector control transfectants, and three fibulin-1D-expressing clones were allowed to grow exponentially and the number of cells was determined at several time points (Table 2). For SHAC cell line and its clonal derivatives, there was no difference in the growth rate of the vector- transfected control cell strains and that of the fibulin-1D-expressing cell strains. For cell line 6AISB1 and its clonal derivatives, introduction of fibulin-1D had no consistent effect on the growth rate of these cells. The three fibulin-1D expressing clones grew somewhat more slowly (23.8-25.8 h per doubling) than the parental cells (20.2 h). One of the vector control clones, CON3, also grew at this slower rate (25.7 h), but did not exhibit a longer than normal tumor latency period. Therefore, the lengthened latency period seen in Figure 7 cannot be accounted for by a slower growth rate of the fibulin- 1D-transfectant clones. 98 Fig. 4. Western blot analysis of the expression and secretion of flbulin-1D protein by Fibulin-1D transfectants. Whole cell lysates and conditioned medium from (A) LG1ceIIs and 6AISB1 cells, three vector-transfected controls (CON1, CON2 and CON3) and six fibulin-1D-expressing transfectants of 6AISB1 cells and (B) SHAC cells, two vector- transfected controls (8a1 and 8b2) and five fibulin-1D-expressing transfectants of SHAC cells were assayed. Cellular protein (20 pg) (upper panel) and 50 pl of the media conditioned by these cell lines (lower panel) were subjected to immunoblotting analysis as described in Figure 3. An arrow indicates the dimer of fibulin-1D, and an arrowhead indicates the monomer. 99 as “E 2P9 909 £39 398 um ONHS 4.9“. v 8...- all QOI GB 99 Oz 92 SNOO ZNOO INOO ISSNB l91 iloxpm .onom .leoww .IDXmBN 100 Fig. 5. Colony formation by fibulin-ID transfected cell strains in 0.33% agarose. Cell strains used were: 6AISB1 cells (A), its vector-transfected control clone CON3 (B) and its fibulin-1D-expressing clone 2b (C) and clone 11a (D), SHAC cells (E), its vector- transfected control clone 8a1(F) and its flbulin-1D-expressing clone 5a3 (G) and clone 5C4 (H). For each cell strain 2 x 104 cells were plated in agarose at 5,000 cells per 60 mm-diameter dish. After 2 weeks (AD) or 3 weeks (E-G) of incubation, a photomicrograph of a representative field was taken from each dish. Representative examples are shown. IOI Fig.5 102 Table 2 Growth properties of parental cells, cells expressing a transfected-fibulin-1 D gene, and vector-transfected control cells Cell Number of colonies of Population Cell No- Of COIONIBS Population Strains a given size per 5,000 doubling strains with a diameter doubling or lines cells assayed in agarose time (h) or lines 2 100pm per time (h) mean 1 SD 5,000 cells mean 1 SD assayed Diameter Diameter 2 150 pm _>_ 200 pm 6AISB1 1250 800 16.7 1 2.1 SHAC 1000 18.4 1 0.5 CON1 1440 730 16.6 1 1.6 8a1 1280 23.2 1 2.1 CON2 1300 700 ND‘ 8b2 1920 22.9 1 2.2 CON3 930 740 23.5 1 4.7 5a3 30 22.1 1 1.1 2b 130 0 27.3 1 4.6 504 0 19.5 1 1.8 2c 100 0 ND 5d2 30 22.6 1 2.0 5c 60 0 ND 5e1 60 ND 8b 990 290 24.1 1 2.4 5f3 0 ND 10b 210 0 ND 11a 30 0 23.2 1 2.2 ‘ Not determined. 103 DISCUSSION In the present study, using mRNA differentially display, we found that the low expression of an extracellular matrix and glycoplasma protein, fibulin-1 D, is associated with malignant transformation. High levels of fibulin-1 expression have been reported in a wide spectrum of normal human tissues and organs (Zhang et al., 1994). In the experiments described here, we assayed 15 tumor cell lines derived from human cancer patients and found that 12 cell lines showed very low levels of fibulin-1 expression. Since the normal cells from which these tumor cells were derived are not available, we cannot be certain that the low expression is the result of downregulation. However, we observed that seven out of 16 MSU-1.1-derived tumor cell lines exhibited markedly reduced levels of fibulin-1 expression compared with the parental MSU-1.1 cells, indicating that down regulation of fibulin-1 expression occurs relatively frequently in the transformation of human fibroblasts in culture. Our data also showed that fibulin-1D downregulation is not specifically correlated with the transformation induced by particular oncogenes or by specific carcinogens. Southern blotting analysis revealed that the downregulation of fibulin-1D in the MSU-1.1 cell strain derivatives cannot be attributed to gross genetic deletions or rearrangements (data not shown). Fibulin-1, together with fibulin-2, belongs to a family of extracellular matrix proteins (Pan 9} al., 1993a; Tran e1 al., 1997). Alternative splicing of fibulin-1 precursor RNA results in four transcripts that encode polypeptides differing from each other only at the carboxyl terminal regions. The four isoforms are designated AD. The dominant forms expressed in most human tissues and cell lines in culture are fibulin-1C and fibulin-1D (Zhang etaL, 1994; Tran et al., 1997). Fibulin-1 proteins are multimodular proteins containing three cysteine-rich anaphylatoxin-related segments, nine epidermal growth factor-like repeats with a consensus motif for calcium-binding, and a variable carboxyl- 104 Fig. 6. In vitro invasiveness of fibulin-1D transfected cell strains in matrigel assays. The invasive index for each cell strain was determined from the number of cells that passed through the matrigel inserts as a fraction of the number of cells that passed through the control uncoated filters. The invasion index of SHAC and 6AISB1 cells was normalized to 100. 105 :o_mo>:_ Egan: 103 11A 23 SC BA CON1 CON2 CON3 2C :o.»u>:_ .3322 513 362 5.3 504 5d2 5.1 SHAC 8'1 6 Fig 106 terminal domain (Argraves et al., 1990; Pan et al., 1993b). Biochemical studies have shown that fibulin-1 interacts with itself and binds other extracellular matrix proteins, including fibronectin (Balbona et al., 1992), laminin, and nidogen (Pan e1 al., 1993b). lmmunohistochemical studies have shown that fibulin-1 proteins are widely expressed intercellular components of connective tissues which are present in matrix fibers and basement membranes (Roark at al., 1995). However, until now little was known about fibulin-1 playing a role in the process of transformation. Since we found that the expression of fibulin-1 was low in many tumor-derived cell lines, we sought to determine whether this low expression of fibulin-l is merely a consequence of malignant transformation, or whether it plays a role in the malignant transformation process. Our results with a transfected fibulin-1 D gene demonstrate that in each case (11 out of 11), elevated expression of fibulin-1 can suppress the ability of fibrosarcoma-derived cells to proliferate in soft agar and to delay the tumorigenicity in vivo. The mechanisms underlying the observed effects have not yet been elucidated. However, a number of studies regarding the role of other ECM molecules in transformation indicate that there is a good correlation between the loss of specific ECM molecules and the acquisition of transformed phenotypes in vitro and tumorigenicity in vivo. For instance, hybrid cells formed by fusion of mouse melanoma cells and normal mouse fibroblasts are not malignant. However, inhibition of fibronectin synthesis in such hybrids by an antisense strategy causes the cells to become tumorigenic (Steel and Harris, 1989). Conversely, addition of purified fibronectin to Herpes simplex virus- transformed hamster fibroblasts can restore normal morphology and adhesive properties characteristic of normal cells (All at al., 1977). Overexpression of fibronectin in human fibrosarcoma-derived HT1080 cells with increased deposition of fibronectin on the cell surface leads to reduced anchorage-independent growth and tumorigenicity 107 Fig. 7. Tumorigenecity of fibulin-1D transfected cell strains in athymic mice. The cell strains used were: SHAC cells (A), 6AISB1 cells (B), their control vector or fibulin-1D transfectants. For each cell line, two mice (total four sites) were injected with 1 x 10° cells at each site. Each symbol represents the volume averaged from four tumors from two mice: parental cell lines (-), control vector transfectants (other closed symbols), and fibulin-1Dtransfectants (open symbols). Average volume of tumors (mma) Average volume of tumors (mm’) 1 200 108 1000 a 800- 600- 400- 200- 0 ' ‘ " T r r 404448 Days after injection (day) O V V O , . , e / l O ‘ A I O é!”t’: _A A . 121620242832364044 Days after Injection (day) Fig. 7 109 (Akamatsu e1 al., 1996). A recent study shows that expression of SPARC, an ECM protein, in ovarian cancer cells results in a significant decrease in cancer cell growth and tumorigenic potential (Mok et al., 1996). Collectively, these data demonstrate that certain ECM molecules play an important role in the regulation of malignant transformation. Perturbation in expression of these molecules could lead to global effects by interfering with the formation and stabilization of extracellular matrix structures, or as suggested by Juliano and Haskill (1993), with normal cell growth signaling pathways that act through extracellular matrix. An important conclusion that can be drawn from our results is that loss of fibulin-1D plays a role in invasion and perhaps metastasis. One possible model is that fibulin-1D negatively regulates the ability of cells to interact with migration-promoting ECM components, thereby inhibiting cellular migration. The ability of fibulin-1 to interact with several adhesive and migration-promoting proteins such as fibronectin and laminin has been established (Balbona et al., 1992; Pan et al., 1993b). It is possible that fibulin-1 binding to these proteins interferes with the ability of cells to derive migratory stimuli. We know that the fibulin-1 binding site in fibronectin is located within type III repeats 13 and 14 (Balbona e1 al., 1992). This region lies adjacent to the Arg-Gly-Asp (RGD)- containing type lllm adhesive domain and the CSIII adhesive domain (Argraves and Gehlsen, 1991). These domains mediate cellular interaction with fibronectin via a wide array of integrins, including 0:581 binding to the RGD site and (1481 binding to the CSIII site. It is possible that the binding of fibulin-1 to fibronectin interferes with integrin binding to either or both of these sites. The fibulin-1 binding site with Engelbreth-Holm- Swarm tumor laminin has been mapped to a site contained within the E3 fragment (Pan et al., 1993b; Brown et al., 1994), a fragment derived from the carboxy terminus of the a chain. This domain has been shown to mediate integrin 0:381 binding (Gehlsen et al., 110 1989; Gehlsen et al., 1992) and lies in the vicinity of binding sites for the integrins (1681 (Aumailley et al., 1990; Hall et al., 1990; Sonnenberg et al., 1991) and 0:781 (Kramer et al., 1991; von der Mark e1 al., 1991). In addition, the laminin receptor dystroglycan binds to the E3 fragment (Gee et al., 1993). Studies are under way to determine whether fibulin-1 modulates any of these receptor-adhesive protein interactions. Interestingly, in contrast to our findings, Clinton at a]. (1996) recently reported that estrogens increase the expression of fibulin-1 in human ovarian cancer cells. They suggest that the augmented fibulin-I expression facilitates ovarian tumor cell invasion. However, they have not yet carried out studies to test this hypothesis. It may also be that estrogen-augmented fibulin-1 expression acts to inhibit tumor cell migration. Clearly, additional experiments are required to determine the role that fibulin-1 plays in the movement and growth of various types of tumor cells. In summary, by using differential mRNA display we have identified fibulin-1 as a gene which is downregulated in a human fibroblastic cell strain transformed in culture. We have found that the fibulin-1D protein is expressed at a low or completely undetectable level in a number of human fibroblastic cells transformed in culture as well as in a number of human tumor-derived cell lines. We have demonstrated that increased expression of fibulin-1D from a transfected gene in human fibrosarcoma-derived cell lines reduced anchorage-independent growth and delayed tumor growth in athymic mice. Furthermore, the invasive ability of these cells was greatly suppressed. These findings indicate that the loss of fibulin-1D expression contributes to the transformation and progression progress of human fibrosarcoma. This study points to the importance of recent studies that have shown that ECM molecules can directly regulate critical cellular process such as growth, differentiation and apoptosis (Jones et al., 1993; Lin and Bissell, 1993; Meredith et al., 1993; Frisch and Francis, 1994). Further research in 111 various types of cells will be required to address the precise mechanism by which ECM contributes to the transformation process. 1 12 MATERIALS AND METHODS Cell§_and_c§ch_ulture The infinite life span human fibroblast cell strain MSU-1.1 and its derivative cell lines were routinely cultured in Eagle's minimum essential medium, modified by addition of L- aspartic acid (0.2 mM), L-serine (0.2 mM) and pyruvate (1mM) and supplemented with 10% supplemented calf serum (803) (Hyclone Laboratory, Logan, UT), penicillin (100 units/ml), streptomycin (100 pg/ml) and hydrocortisone (1 pglml) (complete medium) at 37°C in a humidified incubator containing 5% 002 in air. Qumtluate. Growth curves were obtained by plating 2 x 104 cells per 60 mm diameter dish and harvesting triplicate dishes on day 1, 3, 4, 5 and 7. The total number of cells per dish was determined using an Elzone Counter. The exponential growth phase of the growth curve was used to calculate the population doubling time. Non-tumorigenic human fibroblast cell strain MSU-1.1 and its malignant derivative cell line 6AISB1 were used as sources of RNA. Total RNA from cells in exponential growth was extracted using RNAzolB (Tel-Test, Friendswook, TX). Differential mRNA display was performed as described (Liang and Pardee, 1992; Liang 9.1 al., 1993) with slight modifications. Briefly, total RNA (0.2 pg) from each cell line was reverse transcribed with each of the four degenerate oligo (dT) primers T12MA, T12MT, T12MG and T12MC (where M may be dG, dC, or dA), followed by PCR amplification of the cDNA in the presence of [Oi-35$] dATP (Dupont, Wilmington, DE) using the corresponding T12MN as the 3' primer and one of the arbitrary 10-mers (OPA1-20, Operon Technologies, Alameda, CA) as the 5' primer. The PCR thermal cycle parameters were as follows: 94°C for 30 s, 40°C for 2 113 min, and 72°C for 30 s for 40 cycles, followed by extension at 72°C for 7 min. PCR products were separated in a 6% denaturing polyacrylamide gel. Gels were dried without fixation and exposed to Kodak XAR film. To confirm the results, reactions showing differentially expressed mRNAs in MSU-1.1 and 6AISB1 cells were repeated in duplicate using two independent RNA preparations. The cDNA fragments representing uniquely expressed mRNAs were excised from the dried gels and reamplified by PCR using the same set of primers originally used. The PCR products were run on a 1.5% agarose gel, and the bands of the appropriate size were cut from the gel and purified using QlAquick gel extraction kit (Qiagen, Chatsworth, CA). S | | . | El I? . The purified DNA was used as probes for Northern blot analysis or subcloned into the pCRll vector by the TA cloning method (lnvitrogen, San Diego, CA) according to manufacturer's instructions. Subsequent identification of insert-containing clones was carried out by the dot-blot DNA hybridization procedure described by Callard e1 al.(1994). The subcloned cDNA inserts were again used as probes for Northern analysis to confirm the differential expression. DNA sequencing was performed directly from the TA cloning vector with the SP6 and the T7 primer using the Fidelity DNA sequencing system (Oncor, Gaithersburg, MD). DNA database searches were performed using the GCG FASTA program (Genetics Computer Group, Madison, WI) or the BLAST program from National Center for Biotechnology lnforrnation. Nommhlotjnallsls Total RNA (15 pg) was electrophoresed on a denaturing formaldehyde agarose gel (1.2%), transferred to Hybond-N membrane (Amersham, Arlington Heights, IL) by the downward capillary transfer technique (Chemczynski, 1992) using a 20 fold concentration of standard saline citrate buffer, and fixed by UV crosslinking (UV 114 Stratalinker 2400, Stratagene, La Jolla, CA). DNA probes were radiolabeled by the random primed labeling method (Feinberg and Vogelstein, 1983). Northern hybridization was performed at 42°C overnight in 50% formamide containing 5 x SSPE, 5 x Denhardt’s solution, 0.1% sodium dodecyl sulphate (SDS) and 0.1 mglml salmon sperm DNA. The blots were then washed twice in 1 x SSPE, 0.1% SDS at 42°C, and a third wash was carried out in 0.25 x SSPE with 0.1 % SDS at 55°C. To strip the membrane for reprobing, the blots were treated with boiling 0.1% SDS solution and allowed to cool to room temperature. Variation in RNA loading per lane was evaluated by probing with glyceraldehyde 3-phosphate dehydrogenase (GAPDH) cDNA as the control. E l' E || | | | IT I I. Cells were grown in 100 mm-diameter dishes in Eagles’s medium containing 10% SCS until subconfiuent. To harvest the cells, the cell monolayer was quickly washed four times with cold phosphate buffered saline (PBS), and the cells were lysed for 20 min on ice in 0.5 ml of RIPA buffer composed of 50 mM Tris-HCI, pH 7.2, 150 mM NaCl, 1% Triton X-100, 0.1% SDS, 0.5% deoxycholic acid, 2 mM phenylmethylsulfonyl fluoride (PMSF), 1 mM EDTA and 0.15 units/ml aprotinin. The cell lysates were collected with a rubber scraper, centrifuged at 14,000 x g for 15 min at 4°C, and the protein concentration of the supernatant was determined using a bicinchoninic acid protein assay kit (Pierce Chemical, Rockford, IL) with bovine serum albumin as the protein standard. The cell lysates were used immediately or stored at -80°C until use. To collect conditioned medium, nearly confluent cells cultured in 60 mm-diameter dishes (i.e., ~ 2 x 106 cells) were washed four times with Eagle’s medium, and incubated for another 48 h in 4 ml of complete medium without serum. The conditioned medium was collected, and PMSF (final concentration, 1 mM) and aprotinin (0.15 units/ml) were 115 added, and the medium was centrifuged at 14,000 x g for 5 min to remove cell debris. A second duplicate dish for each sample was used to prepare cell lysate as described above, and the protein concentration of the cell extract was used to normalize the volume of conditioned medium used for Western blot analysis. Aliquots of cell lysates containing 20 pg of protein or 50 pl of the conditioned medium were mixed with the sample buffer (0.05M Tris-HCl, pH6.9, 9% glycerol, 2.3% SDS and 0.1% bromophenol blue) without a reducing agent, and the proteins were separated on a 10% SDS/polyacrylamide gel. The proteins were then electroblotted onto an Immobilion-P membrane (Millipore, Bedford, MA). The blots were blocked for 2 h at room temperature in Tris-buffered saline (20 mM Tris-HCI, pH 7.6, 137 mM NaCI) containing 0.1% (v/v) Tween 20 and 5% (w/v) non-fat dry milk (blocking solution), and then incubated for 2 h at room temperature with the mouse monoclonal anti-fibulin-1 lgG 3A11 diluted 1:20,000 in the same solution. This antibody was generated as part of an earlier study (Argraves at al., 1990). The blots were washed several times and then incubated with horseradish peroxidase-conjugated goat-anti-mouse lgG (Boehringer Mannheim, Indianapolis, IN) that had been diluted 1:3000 with blocking solution. Enhanced chemiluminescence (Amersham, Arlington Heights, IL) was used according to the manufacturer’s recommendations to detect the signal. A 2.3-kb EcoRl fragment of pBluescript-Fibulin-1 D, containing the entire coding sequence of human Fibulin-1 D, was inserted into the unique EcoRl cloning site of the mammalian expression vector pBABE-Puro (Morgenstem and Land, 1990) under the control of Moloney murine leukemia virus long terminal repeat, generating the plasmid pPuro-Fibulin1D. The sense orientation of the cloned fragment was confirmed by 116 restriction mapping. To produce stably transfected cell lines, cells in exponential growth were plated in 100-mm dishes (2 x 105 cells/dish). After 18 to 24 h, 2 pg of plasmid DNA was transfected into the cells using lipofectamine (GIBCO, Gaithersburg, MD) according to the manufacturer’s instructions. Transfected cells were selectively grown in culture medium containing 0.5 pg/ml puromycin (Sigma, St. Louis, M0) for 2-3 weeks. Drug-resistant clones were randomly selected and subcloned individually for further study. WW! For each cell line or strain, 2 x 104 cells were plated at 5,000 cells per 60 mm- diameter dish in a top layer of 0.33% agarose in McM medium containing 2% fetal calf serum, essentially as described by Hurlin at a]. (1989). The agarose plates were maintained at 37°C in a humidified incubator with 3% C02 in air. Each week the medium over the top layer of agarose was removed and fresh McM medium with 2% fetal calf serum was added. At the end of 14 days, 6AISB1 cells and their transfectants or 21 days for SHAC cells and their transfectants, the cells were fixed with 2.5% glutaraldehyde. Photomicrographs covering 0.7 cm2 were made of representative fields from each dish. All colonies greater than 100 lm in diameter were scored and the size of each colony was recorded. The frequency of colonies of a given size in this area from each dish was averaged, and the data were expressed per 5,000 cells assayed. The invasiveness of SHAC cells, 6AISB1 cells and their transfectants was assayed by a modified Boyden Chamber Matrigel method (Albini at al., 1987) according to the manufacturer’s instructions (Collaborative Biomedical Product-Becton Dickson, Bedford, MA). The cells to be studied were washed with PBS three times and harvested by short exposure to 5 mM EDTA. Cells were washed once with Eagle’s medium, and 7 x 105 117 cells in Eagle’s medium containing 0.1 % BSA were seeded onto the control uncoated filters (8 pm pore size) or matrigel-coated filters in Biocoat Matrigel invasion chambers. Eagle’s medium (2.5 ml) containing 0.1% BSA and 10 nglml platelet-derived growth factor (Sigma) was added to the lower compartment. After incubation at 37°C for 48 h (for 6AISB1 cells and its transfectants) or 72 h (for SHAC cells and its transfectants), the filters were fixed in ethanol and stained with 1% crystal violet. Cells that invaded the lower surface of the filters were counted under a light microscope. Each assay was done in triplicate. BALB/c athymic mice 6 weeks of age were injected with 1 X 10° cells in 0.2 ml of serum-free Eagle’s medium subcutaneously in the left front and right hind flank regions. Tumor dimensions were measured twice weekly using a vemier caliper, and the size of tumors was calculated using the formula for the volume of a hemiellipsoid, the geometric figure most nearly approximating the shape of the tumor: Volume=0.5236 X length X width X height (Rockwell at al., 1972). l 18 ACKNOWLEDGMENTS We thank Dr.Georges Guellaen at Hopital Henri Mondor (Creteil, France) for kindly providing EST clone T19384 and Dr. Richard Schwartz of Michigan State University for kindly providing a plasmid containing the 1.3-kb GAPDH cDNA and the expression vector pBABE-Puro. We thank our colleague Dr. Dong Wei for help with DNA sequencing and helpful discussions during the course of this study. This research was supported by DHHS grant CA60907 from the National Cancer Institute, AG11026 from the National Institute of Aging (J. J. M) and GM42912 from the National Institute of General Medicine (W. S. A). 119 REFERENCES Akamatsu, H, lchihara-Tanaka, K, Ozono, K, Kamiike, W, Matsuda, H and Sekiguchi, K. (1996). mm, 56, 4541-4546. Albini, A, lwamoto, Y, Kleinman, HK, Martin, GR, Aaronson, JM, Kozlowski, JM and McEwan, RN. (1987). W., 47, 3239-3245. Ali, lU, Mautner, V, Lanza, R, and Hynes, R0. (1977). Cell, 11, 115-126. Argraves, WS and Gehlsen, KR. (1991). (alive, 5, 489-492. Argraves, WS, Tran, H, Burgess, WH and Dickerson, K. (1990). .L_Qell_B[QL, 111, 3155-3164. Aumailley, M, Timpl, R and Sonnenberg, A. (1990). Exam, 188, 55-60. Balbona, K, Tran, H, Godyna, S, lngham, KC, Strickland, DK and Argraves, WS. (1992). .1._B£QL_QtLem., 267, 20120-20125. Brown, JC, Wiedemann, H and Timpl, R. (1994). J,_C_e[L§_ci.,107, 329-338. Callard, D, Lescure, B and Mazzolini, L. (1994). BLOIQQLIHL'QUQS, 16, 1096-1103. Chomczynski, P. (1992). mm, 201, 134-139. Clinton, GM, Rougeot, C, Derancourt, J, Roger, P, Defrenne, A, Godyna, S, Argraves, WS and Rochefort, H. (1996). W, 93, 316-320. Fearon, ER and Vogelstein, B. (1990). Cell, 61, 759-767. Feinberg, AP and Vogelstein, B. (1983). Andi/322mm, 132, 6-13. Frisch, S and Francis, H. (1994). J._QQILBLQL. 124, 619-626. Gee, SH, Blacher, RW, Douville, PJ, Provost, PR, Yurchenco, PD and Carbonetto, S. (1993). .L_BL'QL_Qh_em., 268, 14972-80. Gehlsen, KR, Dickerson, K, Argraves, WS, Engvall, E and Ruoslahti, E. (1989). _,L_B£QL £11601, 264, 19034-38. 120 Gehlsen, KR, Sriramarao, P, Furcht, LT and Skubitz, AP. (1992). 142215111. 117, 449- 459. Hall, DE, Reichardt, LF, Crowley, E, Holley, B, Moezzi, H, Sonnenberg, A and Damsky, CH. (1990). .LQQILBLQL 110, 2175-84. Hunter, T. (1991). Qell, 64, 249-270. Hurlin, PJ, Maher, VM and McCormick, JJ. (1989). MAM, 86, 187- 191. Jones, PC, Schmidhauser, C and Bissell, MJ. (1993). W., 3, 137-154. Juliano, RL and Haskill, S. (1993). LQQILBL'QL120, 577-585. Kramer, RH, Vu, MP, Cheng, YF, Ramos, DM, Timpl, R and Waleh, N. (1991). Cell 899.111.. 2, 805-817. Liang, P, Averboukh, L and Pardee, AB. (1993). ALL/gleam, 21, 3269-3275. Liang, P and Pardee, AB. (1992). Science, 257, 967-969. Lin, CG and Bissell, MJ. (1993). EASEB_J., 7, 737-743. Meredith, JE, Fazeli, B and Schwartz, MA. (1993). MQL_B£QL_C_QL4, 953-961. Mok, SC, Chan, WY, Wong, KK, Muto, MG and Berkowitz, RS. (1996). Qacogene, 12, 1895-1901. Morgan, TL, Yang, D, Fry, DG, Hurlin, PJ, Kohler, SK, Maher, VM and McCormick, JJ. (1991). mm 197, 125-136. Morgenstem, JP and Land, H. (1990). MAM. 18, 3587-3596. Pan, TC, Kluge, M, Zhang, R-Z, Mayer, U, Timpl, R and Chu, M-L. (1993b). EuL_,.L Biochem., 215, 733-740. Pan, TC, Sasaki, T, Zhang, R-Z, Fassler, R and Timpl, R. (1993a). J._QelLBi_QL, 123, 1 269-1 277. 121 Roark, EF, Keene, DR, Haudenschild, CC, Godyna, S, Little, CD and Argraves, WS. (1995). J.W. 43, 401-411. Rockwell, SC, Kallaman, RF and Fajardo, L. (1972). W., 49, 735-747. Sasaki, T, Mann, K, Murphy, G, Chu, ML and Timpl, R. (1996). W, 240, 427-434. Sonnenberg, A, Gehlsen, KR, Aumailley, M and Timpl, R. (1991). W, 197, 234-244. Steel, DM and Harris, H. (1989). W, 93, 515-524. Tran, H, Mattei, M, Godyna, S and Argraves, WS. (1997). Matulfliology, in press. Von der mark, H, Durr, J, Sonnenberg, A, von der Mark, K, Deutzmann, R and Goodman, S. L. (1991). W., 266, 23593-601. Weinberg, RA. (1989). QanceLBes“ 49, 3713-3721. \NilSOfI, DM, Yang, DJ, Dillberger, JE, Dietrich, SE, Maher,VM and McCormick, J J. (1990). W, 50, 5587-5593. Yang, D, Louden, C, Reinhold, DS, Kohler, SK, Maher, VM and McCormick, JJ. (1992). MW 89, 2237-2241. Zhang, RZ, Pan, TC, Zhang, ZY, Mattel, MG, Timpl, R and Chu, ML. (1994). Genomics, 22, 425-430. CHAPTER III Cloning and Characterization of a Novel Gene Encoding a Putative Transmembrane Protein with Altered Expression in Human Tumors Jing Qing, Dong Wei, Veronica M. Maher, J. Justin McCormick Carcinogenesis Laboratory, Fee Hall Department of Microbiology and Department of Biochemistry The Cancer Center Michigan State University, East Lansing, MI 48824-1316 122 123 ABSTRACT Identification and characterization of genes expressed in normal cells and decreased in their malignant counterparts is an important method for detecting candidate tumor suppressors. Using differential display of mRNAs from nontumorigenic infinite life span human fibroblast cell strain MSU-1.1 and an isogenic fibrosarcoma-derived cell line, 6AISB1, which was derived from chemical carcinogen transformed MSU-1.1 cells, we identified a novel gene, ST7, showing a 6-fold lower expression in 6AISB1 cells compared with parental MSU-1.1 cells. Molecular cloning of a near full-length cDNA revealed that the novel gene encodes a putative transmembrane protein composed of 859 amino acids: the 492 N-terminal amino acids including a 5-fold cysteine-rich repeat of 40 amino acids homologous to the ligand binding repeat of the known low density lipoprotein receptor, a 24 hydrophobic amino acid stretch spanning the plasma membrane, and a C-terminal domain of 343 residues. The ST7 gene is widely expressed in normal human tissues and is particularly abundant in human heart and skeletal muscle. Northern analysis showed that 10 out of 15 tumor cell lines derived from patients and 6 out of 16 cell lines established from tumors formed in athymic mice by MSU-1.1 cells transformed in culture have low or undetectable levels of ST7 mRNA. Furthermore, Western blotting analysis using a specific anti-peptide antibody demonstrated that the levels of ST7 protein are high in normal fibroblasts and low in 12 sarcoma-derived cell lines tested. Altered expression of ST7 appears to occur at both the transcriptional and posttranscriptional level. These studies make it possible to characterize a novel putative receptor protein, whose expression is downregulated in many malignantly transformed fibroblastic cells, and which may play an important role in the transformation of these cells. 124 INTRODUCTION Cancer results from the accumulation of a series of genetic and biochemical changes in normal cells (Peto, 1977; Farber, 1984; Klein and Klein 1985; Fearon and Vogelstein, 1990). Despite the vast increase in our knowledge of oncogenes and tumor suppressor genes associated with human neoplasia over the past 15 years, the molecular events leading to the formation of most types of human tumors are still not well understood. Therefore, identification of genes that are involved in the transition of non-tumorigenic cells to malignant cells can be expected to help us understand the molecular mechanisms underlying tumor formation. The recent introduction of several in vitro transformation systems utilizing human cells in culture (Stoner at al., 1991; Reznikoff e1 al., 1993; McCormick and Maher, 1994; Rhim et al., 1994; Park et al., 1995) has facilitated the investigation of the cellular and molecular mechanisms involved in the multistep carcinogenic process. In our laboratory, transfection of the v-myc oncogene into a human neonatal foreskin-derived fibroblast cell line LG1 led to the establishment of a near-diploid, karyotypically stable, infinite life span human fibroblast cell strain MSU-1.1 (Morgan e_t al., 1991). MSU-1.1 cells are phenotypically normal and do not form tumors in athymic mice. Treatment of MSU-1.1 cells with chemical carcinogens such as benzo(a)pyrene diol epoxide (BPDE) (Yang et al., 1992) or gamma irradiation (Reinhold e_t al., 1996), followed by selection of focus-forming cells, results in cells capable of forming tumors in athymic mice. Since the focus-derived, tumorigenic cells have various properties not possessed by the parental cells, it is presumed that additional genetic alterations induced by carcinogens are required in the transformation process. However, the cellular genes responsible for the neoplastic transformation induced by these carcinogens remain poorly understood. 125 The present study was carried out seeking to isolate the oncogene(s), or tumor suppressor gene(s) that are involved in this neoplastic conversion. The recently developed differential mRNA display method (Liang and Pardee, 1992; Liang at al., 1993) allows one to identify genes that are differentially expressed between closely related eukaryotic cells. In our study, we applied this method to compare the mRNA profile between the nontumorigenic parental MSU-1.1 cells and one of its malignant derivative cell lines, 6AISB1, which was established from a fibrosarcoma formed in an athymic mouse by BPDE-transformed MSU-1.1 cells. We have identified a novel gene, designated ST7, whose mRNA is markedly downregulated in 6AISB1 cells compared with MSU-1.1 cells. A near full-length cDNA was cloned and the deduced amino acids revealed that this novel gene encoded a putative transmembrane protein of 859 amino acids. ST7 mRNA and protein levels were low in a large fraction of the tumor derived cell lines tested. Further characterization of ST7 should lead to better understanding of the carcinogenesis process in human cells. 126 RESULTS ldentlfleatien Qf $11 as a Gene Differentially Examssed BeMeen W and Malignant Human Calls. When we compared MSU-1.1 cell strain and its tumorigenic derivative cell line 6AISB1 using mRNA differential display, we saw 8,000-10,000 displayed cDNA fragments and found in two independent experiments that nine DNA fragments reproducibly showed differential intensities between MSU-1.1 cells and the tumor-derived cells (Figure 1A and data not shown). cDNA fragments were isolated from the sequencing gel and used as probes in Northern analysis to test for differential mRNA expression between MSU-1.1 and 6AISB1 cells. Differential expression of mRNA was confirmed for five of the nine cDNAs. Subsequent analysis showed that four of these five corresponded to the same gene, i.e., fibulin-1D (Qing at al., 1997). The fifth, designated ST7, is shown in Figure 1B. This cDNA fragment hybridized with a 3.7 kb transcript showing a 6-fold lower expression in 6AISB1 cells than in MSU-1.1 cells. The ST7 DNA fragment was subcloned, and the insert DNA from several individual plasmids was also found to hybridize to the same sized RNA and the transcript exhibited differential expression in MSU-1.1 cells compared with the tumor cell line. Subsequent sequencing analysis of this partial cDNA revealed that it contained 485 hp with no significant homology with any known genes in the nucleotide sequence databases. Expression $21 $11 in Multiple Human Tumor Cell Lines. To test whether the expression of ST7 mRNA is also downregulated in other tumor-derived MSU-1.1 cell lines malignantly transformed by various methods, RNA from 15 additional such cell lines was assayed by Northern blotting analysis. We also examined 15 tumor-derived cell lines from patients. The origin of these cell lines has been described previously (Qing at al., 1997). Representative data are shown in Figure 2. We found 127 downregulation of ST7 in 5 out of 15 MSU-1.1 derivatives (Lanes 4, 5, 8, 21 and 22). We also found that in 10 out of 15 tumor-derived cell lines from patients, the expression of ST7 was very low or undetectable, i.e., in cells from three out of five fibrosarcomas (Lanes 10, 12-13), in cells from two out of two osteosarcomas (Lanes 6 and 16), in cells from the three neurofibrosarcomas (Lanes 18-20), and in cells from a cervical carcinoma (Lane 7) and a bladder carcinoma (Lane 3). Collectively, of the 31 tumor-derived cell lines assayed, 16 exhibited either low or no expression of ST7. Meleeular filming of the Eullzlengtn Human SIZ cDNA. To obtain the full-length cDNA of ST7, we screened a human fibroblast cDNA library (library 9 of Legerski) with an ST7 gene-specific primer and a vector-specific primer, using the High Fidelity Expand polymerase chain reaction method (Boehringer Mannheim). Several specific PCR products were obtained. We sequenced the longest one (clone A), which was about 2.6 kb (Figure 3A). To recover the 5' end of the cDNA, we screened a human skeletal muscle cDNA library with another ST7-specific primer and a vector primer using the PCR method described above. This produced a fragment we called clone B. To obtain additional 5' sequence, we carried out the 5'-rapid amplification of cDNA ends (RACE) reaction using a human heart Marathon-ready cDNA (Clontech, Palo Alto, CA) and designated the generated fragment clone C. These three clones overlapped with each other (Figure 3A). Two forms of the cDNA differing from each other at the 5' end were isolated. The longer form has 57 more nucleotides than the shorter one. The assembled nucleotide sequence of the longer form (total 3078 bp) and the deduced amino acid sequence are shown in Figure 3B. An open reading frame extends from nucleotide 43 to 2619 and encodes a protein of 859 amino acids with an estimated molecular weight of 92.8 kDa. The first ATG codon (nucleotides 43-45) lies in the context of the Kozak 128 Fig.1. (A) Differential display comparing mRNAs from the parental MSU-1.1cell strain (lanes 1 and 2) and its tumorigenic derivative cell line 6AISB1 (lanes 3 and 4). For each cell line, two independent preparations of total RNA were extracted, reverse transcribed, and amplified by PCR in the presence of [or-3°SldATP. The PCR products were separated on a 6% polyacrylamide gel and autoradiographed. The signal demonstrating altered expression is marked by an arrow. The primers used were T12MT and OPA7 as described by Qing at a]. (1997). (B) Northern blot analysis confirming differential gene expression for fragment ST7 using the amplified DNA fragment as a probe. Total RNA (15 pg) obtained from MSU-1.1 cells (lane 1) and 6AISB1 cells (lane 2) was subjected to Northern analysis as detailed in “Materials and Methods”. The blot was stripped and reprobed with a glyceraldehyde 3-phosphate dehydrogenase (GAPDH) cDNA (lower panel) as the loading control. 129 .0 .‘h it I : «ST7 N If!“ |l . 1'ST7 ..<-GAPDH 130 consensus initiation site of eukaryotic mRNA translation (Kozak, 1991). The $17 cDNA contains a putative 3' untranslated region of 450 bp followed by a poly A tail and three consensus polyadenylation signals (Wahle and Keller, 1992) at nucleotide positions 2846, 2976 and 3034. Comparison of $17 cDNA sequence to the Genbank and EMBO database using the FASTA and the BLAST program revealed that there is no significant homology with any known genes. Enmanr StmfluLe Suggest: a Nolel Itanemembtane ELotein. Comparison of the deduced amino acid sequence of ST7 with a nonredundant protein sequence database revealed that overall there was no significant homology with other proteins. A striking feature of this protein is that the amino-tenninus is composed of five imperfect repeats of a 40-amino acid cysteine-rich repetitive sequence homologous to that found in the human low density lipoprotein (LDL) receptor (Shdhof at al., 1985) (Figure 4) and complement protein C9 (Stanley e1 al.. 1985). The similarity also includes the highly conserved, negatively charged Ser-Asp-Glu triad, which occurs near the COOH-terminal end of each repeat. Hydropathy analysis (Kyte and Doolittle, 1982) revealed that the coding sequence contained a stretch of 24 hydrophobic amino acids extending from residues 493 to 516 (underlined in Figure 3B), which is flanked at both ends by positively charged residues. This hydrophobic sequence resembles the membrane- spanning region of other transmembrane proteins (Sabatini at al., 1982). The predicted protein also contains other putative functional domains, including nine putative N-linked glycosylation sites, a number of potential phosphorylation sites for protein kinase C and casein kinase II, and several N-myristoylation sites. The importance of these putative domains of ST7 protein under physiological conditions remains to be determined. 131 Fig. 2. Northern blot analysis of ST7 expression in multiple human tumor cell lines. (A) Total RNA (15 pg), obtained from a series of cell lines, was hybridized with ST7 cDNA using a cloned ST7 DNA fragment as a probe. These were: fibroblast cell line from a normal donor (lane 1); parental MSU-1.1 cells (lanes 2 and 9); malignant cell lines derived from a patient’s cervical carcinoma (lane 3); malignant cell lines derived from tumors formed in athymic mice alter injection of MSU-1.1 cells transformed by a transfected oncogene (lane 4) or by carcinogen treatment (lane 5); malignant cell lines derived from a patient’s osteosarcoma (lanes 6 and 16), bladder carcinoma (lane 7); malignant cell lines derived from tumors formed in athymic mice after injection of MSU- 1.1 cells spontaneously transformed (lane 8); malignant cell lines derived from a patient’s fibrosarcoma (lanes 10-14), rhabdomyosarcoma (lane 15), Wilms' tumor (lane 17), and neurofibrosarcoma (lanes 18-20); and malignant cell lines derived from tumors formed in athymic mice after injection of MSU-1.2 cells transformed by cobalt 60 (lanes 21 and 22). (B) RNA loading, evaluated by rehybridizing the same blots with a GAPDH cDNA probe. ZJ-Z'l'nSlN IJ-Z'l-DSW Luunsm LIJIfISM lull-:IN eoeewo 9931 a a oeour-r L888 OVHS .l.:I=dIA ION l'I-I‘ISIN JJ./IQV'I 173 .I. M188“- 183/0” 10 2'82 1- Iflat-I l' L'DSW 6818 < 132 b. on: “ 910111213141516171819 202122 2345678 1 133 Fig. 3. Characterization of ST7 cDNA clones. (A) A diagram showing the four overlapping ST7 cDNA clones. They were isolated from MSU-1.1 cells, a human fibroblast library, a skeletal muscle cDNA library, and a human heart Marathon-ready cDNA, as described in Materials and methods. (B) A near full-length cDNA sequence of ST7 gene and the deduced amino acid sequence. The amino acid sequence is numbered beginning with the first methionine of the longest open reading frame. The one-letter code is used for amino acids. The 19 amino acids existing only in the longer isoform is indicated by double underlining. The putative transmembrane domain is underlined. The peptide used for antibody generation is in bold font. The putative polyadenylation signals are indicated with sets of asterisks. 134 A MSU-1.1 [:1 oopcn (0.5 kb) Library 9 l 1 Clone A (2.8 kb) ' Skeletal Muscle [:3 Clone B (0.7 kb) Normal Heart :3 Clone C (0.7 Kb) 135 CTCCTCCTCCGTCTCCTCCTCTCTCTCTCATCTGCTGTGGTTATGGCCTGTCGCTGGAGCACAAAAGAGTCTCCGCGGTGGAGGTCTGCGTTGCTCTTGCTUTCCTCGCTGGGGTGTAC HACRWSTKESPRNRSALLLLFLAGVY (ISMATGGTGCTCTTGCAGAAMTTCTGAAAATGTGCATAUTCAGGAGTGTWCTGCTTGTGGAGAGACTCCAGAGCAAATACGAGOACCAAGTGGCATAATCACAAGCCCAGGCTGG GNQALAEHJENVHJSGVSTACGETPEOIRAPSGIITSPGH CCTT CT GAATATCCTGCAAAAATCAACTGTAGCTGGTT CATAAGGGCAAACCCAGGCGAAATCATTACTATAAGWTTCAGGATTTTGATATTCAAGGATCCAGAAGGTGCAATITGGAC PSEYPAKINCSNFIRANPGEIITISFQDFDIOGSRRCNLD TGGTTGACAATAGAAACATACAAGAATATTGAAAGTTACAGAGCTTGTGGTTCCAOAATTCCACCTCCGTATATCTCTTCACAAGACOACATCTGGATTAGGTTTCATTCGGATGACAAC HLTIETYKNIESYRACGSTIPPPYISSQDHIHIRFHSDDN ATCTCTAGAAAGGGTI'TMGACTGGCATA | l | l I CAGGGAAATCTGAGGAACCAAATTGTGCTTGTGATCAGTFTCGTT GT GGTAATGGAAAGT GT ATACCAGAAGCCTGGAAATGCAAT ISRKGFRLAYFSGKSEEPNCACDOFRCGNGKCIPEAHKCN AACATGGATGAATGTGGAGATAGTTCCGATGAAGAGATCTGTGCOAAAGAAGCAAATCCTCCAACTGCTGCTGCTTTTCAACCCTGTGCTTACAACCAGTTCCAGTGTTTATCCCGTTTT NHDECGDSSDEEICAKEANPPTAAAFQPCAYNQFQCLSRF ACCAAAGI'ITACACTTGCCTCCCCGAATCTTTAAAATGTGATGGGAAOATTGACTGCCTTGACCTAGGAGATGAGATAGACTGTGATGTGCCAACATGTGGGCAATGGCTAAAATATTIT TKVYTCLPESLKCDGNIDCLDLGDEIDCDVPTCGQHLKYF TATGGTACTTTTAATTCTCCCAATTATCCAGACTTTTATCCTCCTGGMGCAATTGCACCTGGTTAATAGACACTGGTGATCACCGTAAAGTCAWTTACGCTTCACTGACTWAAACTT YGTFNSPNYPDFYPPGSNCTHLIDTGDHRKVILRFTDFKL GATGGTACTGGTTATGGTGATTATGTCAAAATATATGATGGATTAGAGGAGAATC CAMCAAGCTWTGCGTGTGTTGACAGCTTTTGATTCTCATGCACCTCTTACAGTTGTTTCTTCT DGTGYGDYVKIYDGLEENPHKLLRVLTAFDSHAPLTVVSS TCTGGAOAGATAAGGGTAUI l l l | GT GCTGATAAAGTGAATGCTGOAGGGGATTTAATGCTACTTACCAAGTAGATGGGTT CT GTTT GCCATGGGAAATACCCT GTGGAGGTAACTGG SGQIRVHFCADKVNAARGFNATYOVDGFCLPHEIPCGGNH GGGTGTT ATACTGAGCAGCAGCGTT GT GATGGGT ATTGGCATTGCCCAAATGGAAGGGATGAAACCAATTGTACCATGT GCCAGAAGMAGAATTTCCATGTT CCCGAAATGGTGT CTGT GCYTEQQRCDGYHHCPNGRDETNCTNCQKEEFPCSRNGVC TATCCTCGTTCTGATCGCTGCAACTACCAGAATCATTGCCCAAATGGCTCAGATGAAAAAAACTGC | | | l | l | GCCAACCAGGAAATTTCCATTGTAAAAACAATCGTTGTGTGTTTGM YPRSDRCNYQNHCPNGSDEKNCFFCQPGNFHCKNNRCVFE AGTT GGGTGT GT GATTCT CAAGATGACTGTGGT GATGGQGCGATGAAGAAAATTGCCCAGT AATCGTGCCTACAAGAGTCATCACTGCTGCCGT CATAGGGAGCCT CATCT GT GGCCTG SHVCDSQDDCGDGSDEENCPVIVPTRVLTAAVIGSLICEL TTACTCGTCATAGCATTGGGATGTACW GTAAGCTTTATTCTCTGAGAATGTTT GAAAGAAGATCATTTGAAAOACAGTT GT CAAGAGT GGAAGOAGAATTGTTAAGMGAGAAGCTCCT L_LV1ALGCTCKLYSLRMFERRSFETOLSRVEAELLRREAP CCCT CGT ATGGACAATTGATTGCT CAGGGTTT AATTCCACCAGTTGAAGATTTTCCT GmGTTCACCT AATflGGCTTCT G I | l IGGAAAATCTGACISCTAGCGGTACGATCTCAGCTT PSYGQLIAOGLIPPVEDFPVCSPNOASVLENLRLAVRSQL GGATTTACTTCAGTCAGGCTTCCTATGGCAGGCAGATCAAGCAACATTTGGAACCGTAI | l | lAATHTGCAAGATGACGTCATTCTGGGTCATTGGCTTTGGTCTCAGCAGATGGAGAT G FTSVRLPIIAGRSSNIHNRIFNFARSRHSGSLALVSADGD GAGGTTGTCCCTAGTCAGAGTACCAGTAGAGAACCTGAGAGAAATCATACTCACAGAAGTTTGTTTTCCGTGGAGTCTGATGATACAGACACAGAAAATGAGAGAAGAGATATGGCAGGA E VVPSQSTSREPERNHTHRSLFSVESDDTDTENERRDHAG GCATCTGGTGGGGTTGCAGCTCCTTTGCCTCAAAAAGTCCCTCCCACAACGGCAGTGGAAGCGACAGTAGGAGCATGTGCAAGTTCCTCAACTCAGAGTACCCGAGGTGGTCATGCAGAT A SGGVAAPLPQKVPPTTAVEATVGACASSSTQSTRGGHAD MTGGAAGGGATGTGACAAGT GT GGAACCCCCAAGTGT GAGTCCAGCACGTCACCAGCTTACAAGTGCACTCAGT CGTATGACT CAGGGGCTACGCTGGGTACGWTTACATTAGGACGA N GRDVTSVEPPSVSPARHQLTSALSRHTQGLRIIVRFTLGR TCAAGTTCCCTAAGTCAGAACCAGAGTCCTTTGAGAMACTTGATAATGGGGTAAGTGGAAGAGAAGATGATGATGATGTTGAAATGCTAATTCCAATITCTGATGGATCTTCAGACTTT S SSLSQNQSPLRQLDNGVSGREDDDDVEI‘ILIPISDGSSDF GATGTGAATGACTGCTCCAGACCTCTTCTTGATCTTGCCTCAGATWGGAWGGGCWAGAWCCATATAATGCMCAAATCCTGGAGTAAGGCCAAGTAATCGAGATGGCCCCTGT D VNDCSRPLLDLASDQGQGLRQPYNATNPGVRPSNRDGPC GAGCGCTGTGGTATTGTCCACACTGCCCAGATAC CAGACACTTGCTTAGAAGTAACACTGAAAAACGAAACGAGTGATGATGAGGCTFTGTTACTTTGTTAGGTACGMTCACATAAGGG E RCGIVHTAQIPDTCLEVTLKNETSDDEALLLC AGATTGTATACAAGTTGGAGCAATATCCATTTATTAUTTGTAACTTTACAGTTMACTAGTTTTAGWTAAAAAGAAAAAATGCAGGGTGATTTCTTATTATTATATGTTAGCCTGCAT GGTTAAATTCGACAACTTGTAACTCTATGAACTTAGAGTTTACTATTTTAGCAGCTAAAAATGCATCACATATTGCATATTGTTCAATAATGGTCCTTTCATTTGTTTCTGATTG| l 1 IC ****** ATCCTGATACTGTAGTTCACTGTAGAAATGTGGCTGCTGAAACTCATTTGATTGTCAl l l I IATCTATCCTATGTTAAATGGI l IGI l | l IACAAAATAATACCTTATTTTAATTGAAAC ****** GTTI'ATGCI | l IGCCAAGCACATCTTGTMCTTAATATAGCTAGATGTTAAGGTTGTTAATGTACCAAAAAMAMAA *iiiit Figure 38 120 26 240 360 106 480 146 600 186 720 226 840 266 960 306 1080 346 1200 386 1320 426 1440 466 1660 506 1680 546 1800 586 1920 626 2040 2160 706 2280 746 2400 786 2520 826 2640 859 2760 2880 3000 3078 136 Banem 9.1 Emmaion 91 SIZ mRNA in Manage Human Iiesues. Northern analysis using a human normal tissue blot (Clontech) revealed that ST7 cDNA hybridized to a single transcript of 3.7 kb. The ST7 mRNA was found to be most abundant in heart and skeletal muscle, expressed at moderate levels in brain, lung, placenta and pancreas, and barely detectable in tissues containing large number of epithelial cells such as liver and kidney (Figure 5). mmmmmflmjflnmflmanlumarmm. Arabbitanti- ST7 polyclonal antibody (designated B250) was raised against a synthetic peptide as described in Materials and Methods. This antibody recognized a protein with an apparent molecular mass of 85 kDa, which is lower than the estimated molecular mass. The reason for this anomalous migration of ST7 protein in SDS-polyacrylamide gel is not clear- The specificity of this antibody for ST7 protein was tested by determining the extent of inhibition with the antigen peptide. After preincubation of the antibody with the antigen peptide (60 uM), the signal for the ST7 protein band in immunoblots was dramatically reduced (Figure 6). Using this antibody, the expression level of ST7 protein in normal cells and tumor-derived cell lines was analyzed by Western blotting analysis. As shown in Figure 7, MSU-1.1 cells and normal human fibroblast cell line SL89 exhibited high levels of $17 protein, whereas the six malignant MSU-1.1 derivatives (lanes 3-8) and three cell lines derived from human fibrosarcomas (lanes 9-11) showed low levels of ST7 protein, which was consistent with the levels of ST7 mRNA transcripts in these cells (Figure 2). Interestingly, human fibrosarcoma-derived cell lines HT1080 and VIP:FT and rhabdomyosarcoma-derived cell line RD showed an mRNA level of ST7 comparable to that in MSU-1.1 cells (Figure 2, compare lane 9 and 11,14 and 15), whereas the protein was undetectable (Figure 7, compare lane 1 and 12-14). This result 137 Fig. 4. Alignment of the cysteine-rich repeat region of ST7 with that of the human LDL receptor. Amino acids are numbered on the left. Conserved amino acids are boxed. The data of human LDL receptor are taken with permission from “The LDL receptor gene: A mosaic of exons shared with different proteins” by Sudhof at al., Science, 1985. 138 U] L) U C) m 0 CT (4 [L5 C) d: 3 Id m 23 3 O X: C) DJ (J L) L) [U m C) m (D C) r0 (J L) U >flz x“. a: I 01 r0 0| C) M [5) I: C) ’Eritu 7h ha ha A (3 C) d U] L) L9 L) o (J o L5) m m m mmvlmvv mevlaav OHvIOBM vaIoHN oomrama .oz osoflmom £995 Cw .m MDMCUWCOU > z > mmNIomN fl > 4 mvNIHHN m m o oamlmha A > m meIMNH x m m NHHrvm > m A mmIMv a o > ~e-~ .02 ODUfimOm 5389-43 .< 139 suggests that downregulation of ST7 gene product occurs at both transcriptional and posttranscriptional levels. 140 Fig. 5. Expression of ST7 gene in normal human tissues. (A) Human multiple tissue blot (Clontech) containing poly (A)+ RNA (2 jig/lane) from the indicated tissues probed with labeled 2.6 kb ST7 cDNA fragment. (B) The blot, stripped and rehybridized with a probe for B-actin as a loading control. 141 mamaocmn. >962 eEms—2 68.3w ..o>_._ was; Scoomi 59m two... (Kb) 142 Fig. 6. Analysis of the specificity of the antibody B250. lmmunoblots of cell lysate (25 pg protein/lane) from MSU-1.1 cells were incubated with the rabbit anti-ST7 peptide antibody B250 without (lane 1) or with (lane 2) prior incubation with the antigen peptide. The position of the ST7 signal is indicated by an arrow. 143 c It“: a. 1 ¢ 85 kDa Fig.6 144 Fig. 7. Western blot analysis of the expression of ST7 in normal human fibroblast cell lines (MSU-1.1 and SL89) and 12 tumor-derived cell lines. Cellular protein (25 pg) was analyzed by electrophoresis in 10% polyacrylamide gel and probed with antibody B250. The origin of the tumor-derived cell lines are: malignant cell lines derived from tumors formed in athymic mice after injection of MSU-1.1 cells transformed by various methods (lanes 3-8); malignant cell lines derived from a patient’s fibrosarcoma (lanes 9-13) and rhabdomyosarcoma (lane 14). 145 n— m tun—5 one 2.... .02 swam U