,P u. 0 .5‘ I unit‘s 3?... . ‘ . . . ~v.......u .5. u. 3.! L. . u a: a. I:.!.1!931.I.l.l.r :2. 3 "u, v [(I1Y 1. I .‘ . {III}... 1| arl’llE...“ t A -‘35‘ I l' 3].! '2 l"! ‘1 I ' , . . . i. u: . . Ola. fissfitazsznu X INA-unfit?” v4.5 .(htnjr. LT ,..r§vt..vl#..t9..v.._. F :2. w d: .. 4...... u. _ . y: .|.,. 2% 1.. . .. , .1... .. AK... HI .AA... ......Tl..: .P‘rr I’h... Ain.“.~w....m..,-__l..r9.s35'. gnu. v'uo . ‘ ‘ s. .. . . . , ‘ SETAT UNIVERSITY LIBRARIES 'llllllll W l liill lli ill 3 1293 008820 This is to certify that the dissertation entitled ONCOGENE COOPERATION AND CELLULAR DIFFERENTIATION IN THE IN VIVO AND IN VITRO TRANSFORMATION OF MURINE PRE-B CELLS presented by Shu-Chih Chen has been accepted towards fulfillment of the requirements for PhoDo degree in MicrObiOlogy '7 t , Major Professor 6/ ( Date March 25, 1992 MS U i: an Affirmatiw Action/Equal Opportunity Institution 0- 12771 _____ _ ,V_ _ . ._ . — A- I, _ A — _ r i ' _ LIBRARY Michigan State '1 University PLACE IN RETURN BOX to remove this checkout from your record. TO AVOID FINES return on or before date due. J DATE DUE DATE DUE DATE DUE l __i J MSU Is An Affirmative ActiorVEqual Opportunity Institution cmmunii-ni ONCOGENE COOPERATION AND CELLULAR DIFFERENTIATION IN THE IN VIVO AND IN VITRO TRANSFORMATION OF MURINE PRE-B CELLS BY Shu-Chih Chen A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Department of Microbiology and Public Health 1 992 (77-a7g'0 ABSTRACT ONCOGENE COOPERATION AND CELLULAR DIFFERENTIATION IN THE IN VIVO AND IN VITRO TRANSFORMATION OF MURINE PRE-B CELLS BY ShU-Chih Chen An in vivo and an in vitro model system were used to study oncogene cooperation which presumably occurs in multi-step tumorigenesis. Both model systems involved transforming murine B cells using the Whitlock and Witte culture system in combination with tumor challenge. In the in vivo model system, c-myc gene activation was found in tumors derived from a v-Ha-ras cell line. Two tumors possessed a MoMuLV provirus integration immediately upstream and in a reverse transcriptional orientation to c—myc. Elevated expression of c-myc was found in these two tumors and another two tumors with no gross gene alteration. This finding parallels the synergy of v—Ha—ras and v—myc observed in the in vitro transformation of murine B lymphoid cells and validates synergy as a model for in vivo tumor progression. The insertional activation of the c-myc gene by MoMuLV in B cell lymphomas is novel. The flanking region, bone-1, of one of these non-c- myc tumor-specific viral integration sites was characterized. Sequence homology to this locus was found in other mammals, and chicken. In the in vitro model system, lL-7, a pre-B cell growth factor, was found to be incapable of cooperating with the v-Ha-ras oncogene in inducing a fully transformed phenotype in murine B cells. A discrepancy between the oncogene cooperation in a co-infection procedure and a sequential addition procedure was found. The clonal nature of the cell lines generated from the coinfection procedure suggests the selection of additional oncogenic events. The oncogenic potential of lL-7 expression, in itself, and those of other genes are probably best assessed in well-characterized individual cell lines. A “lineage switching" from v-Ha-ras transformed pre-B cells to "macrophage- like" cells was also found in this study. These pre-B cells have gained the capacity to effectively present antigen in an MHC-restricted fashion. These cells have also rearranged their kappa light chain immunoglobulin locus, suggesting that macrophage differentiation and immunoglobulin rearrangement are not mutually exclusive processes. The existence of both lymphoid and myeloid characteristics in a cell suggests greater plasticity in hematopoietic lineage commitment than conventionally thought to be the case. TO MY MOM AND DAD iv ACKNOWLEDGMENTS I would like to express my deepest appreciation to my major professor, Dr. Richard Schwartz for his direction, encouragement, and patience during my years as a graduate student. I also would like to thank the members of my committee for their guidance and invaluable time, Drs. Jerry Dodgson, Susan Conrad, Michele Fluck, and Lyman Crittenden. I thank Diane Redenius for her technical assistance and friendship, and James Bretz and Timothy Weichert for their companionship. They and Rich have taught me lots about American culture and made my stay in Michigan State University fun and colorful. Thanks are due the members of Dr. Dodgson’s Laboratory for their generosity in letting me share equipments and materials. Last, but not least, I want to thank my parents Mr. S.C. Chen and Mrs. H.F. Tsai, my sister H.H. Chen, my brothers Y.S. Chen and Y.Y. Chen for their endless love and support. Thanks are to all my friends, especially H.-C. Li, in East Lansing, too. With them, life has been delightful for the past five years. TABLE OF CONTENTS Page List of tables ....................................................................................................... viii List of figures ......................................................................................................... ix Introduction .......................................................................................................... 1 References ................................................................................................ 4 Chapter 1 Literature review 1. Tumorigenesis is a multi-step process ......................................... 5 2.1 The search for genes that are involved in tumorigenesis .............. 8 2.1.1 Identification of cellular oncogenes by DNA transfer experiment..8 2.1.2 Characterization of viral flanking regions ...................................... 11 2.1.3 Molecular characterization of chromosomal aberrations .............. 15 2.1.4 Identification of new oncogenes by homologous recombination..18 2 2 Experimental models for transformation and tumor progression..19 2.2.1 Use of animal model versus human models ................................. 19 2.2.2 Use of in vivo versus in vitro systems .......................................... 20 2.2.3 Model systems used to test the oncogenic potential of oncogenes ..................................................................................... 20 2.2.4 Model systems used to study tumor progression ........................ 22 3. Multiple genes or factors that are involved in B cell lymphomas/leukemias of human, mouse, and avian ................... 24 3.1 Human B cell neoplasia ................................................................ 24 3.1.1 Burkitt lymphoma .......................................................................... 24 3.1.2 Other human B-cell leukemias and lymphomas ............................ 30 B-cell chronic lymphocytic leukemia ............................................. 30 Follicular lymphomas ..................................................................... 33 Pre-B cell acute lymphocytic leukemia .......................................... 34 3.2 Murine plasmacytoma .................................................................... 35 3.3 Avian lymphoid leukosis ................................................................ 39 3.4 In vitro transformation of murine B cells ....................................... 42 Literature cited ......................................................................................... 48 Chapter 2 Tumorigenesis of a v-Ha-ras-expressing pre-B cell line selects for c-myc activation .......................................................................... 61 Abstract .................................................................................................... 61 Materials and methods ............................................................................ 62 Results ..................................................................................................... 62 Discussion ................................................................................................ 66 References ............................................................................................... 67 vi Chapter 3 Cloning and characterization of a viral flanking region common to B lymphoid tumors derived from a v-Ha-ras infected pre-B cell line .................................................... 69 Abstract .................................................................................................... 70 Introduction .............................................................................................. 71 Materials and methods ............................................................................ 73 Results ..................................................................................................... 76 Discussion ................................................................................................ 85 References ............................................................................................... 87 Chapter 4 IL-7 expression in a v-Ha-ras transformed pre-B cell line is not sufficient for tumorigenicity: differing assessments in clonal versus heterogeneous populations ........................................ 89 Abstract ..................................................................................................... 90 Introduction .............................................................................................. 91 Results ..................................................................................................... 93 Discussion ................................................................................................ 107 Materials and methods ........................................................................... 109 Acknowledgement .................................................................................. 1 12 References .............................................................................................. 1 13 Appendix A Lineage switch macrophages can present antigen ......................... 115 Abstract ................................................................................................... 1 15 Introduction ............................................................................................. 1 16 Results .................................................................................................... 1 17 Discussion .............................................................................................. 135 Materials and methods ........................................................................... 136 Acknowledgement .................................................................................. 141 References .............................................................................................. 142 Appendix B ....................................................................................................... 145 Introduction ............................................................................................ 145 Materials and methods ........................................................................... 145 Results .................................................................................................... 147 Discussion .............................................................................................. 159 References .............................................................................................. 162 Summary and discussion .................................................................................. 163 References .............................................................................................. 172 vii LIST OF TABLES Page Table Chapter 1 1.1 Oncogenes identified by DNA transfer experiment ...................................... 10 1.2 Cellular genes activated by proviral insertion ............................................... 12 1.3 Viral flanking regions without viral oncogenes analogues ............................ 13 1.4 Genes identified from chromosomal aberrations .......................................... 17 Chapter 2 1. Restriction fragments in the 5’ c-myc region of tumors 1 and 3 ................... 65 Chapter 4 1. Growth without cellular feeder layers ........................................................... 101 2. Soft agar growth and tumorigenesis ............................................................ 101 Appendix A 1. LPS-induced cytokine released by tumor 4 macrophage subclones .......... 124 Appendix B 1. Tumor challenge in Balb/c mice .................................................................... 150 2. Probes that were used in searching for the putative secondary events that may be involved in the tumor progression of R2 tumors ............... 151 3. Relative stability of c-myc mRNA in R2 and tumors .................................... 155 viii LIST OF FIGURES Page Figure Chapter 2 1. Viral ras integrations ....................................................................................... 63 2. MoMuLV integrations ..................................................................................... 64 3. Rearrangement of c-myc in tumor 1 and 3 .................................................... 64 4. MoMuLV integration near c-myc .................................................................... 65 5. env and c-myc cohybridize ............................................................................ 66 6. Elevated c-myc mRNA levels .......................................................................... 66 Chapter 3 1. Tumor specific viral integration in the bone-1 locus ...................................... 77 2. Cloning stragegy and restriction map of the bone-1 locus ............................ 79 3. Bone-1 is evolutionary conserved .................................................................. 81 4. DNA sequences of the 03P2 fragment .......................................................... 82 5. Putative open reading frames in the sequence of the 03P2 fragment .......... 83 6. RFLP mapping of bone-1 ................................................................................ 84 Chapter 4 1. Proviral integration .......................................................................................... 94 2. Retroviral transcription .................................................................................... 98 3. Immune loci define a pre-B phenotype ......................................................... 100 4. Proviral integration ........................................................................................ 105 5. Retroviral transcription ................................................................................. 106 Appendix A 1. Viral integrations ........................................................................................... 118 2. Nonspecific phagocytosis of latex beads ..................................................... 120 3. RNA analyses of c-myc, c-myb, and c-fms ................................................... 122 4. Kappa light chain rearrangement ................................................................. 125 5. Expression of mu chain RNA ....................................................................... 127 6. RT-PCR analysis of CD45 ............................................................................ 131 7. Antigen presentation .................................................................................... 132 8. l-ACl expression ............................................................................................. 133 Appendix B 1. Growth of R2 and seven tumors in the presence and absence of IL-7 ....... 149 2. Nuclear run-on transcription ........................................................................ 153 3. Kappa chain gene rearrangement ................................................................ 157 4. Mu chain gene rearrangement ..................................................................... 158 Introduction The focus of my thesis has been the identification of putative oncogenes that can cooperate with v-Ha-ras in tumor progression or in the in vitro transformation of murine B lymphoid cells. Two model systems involving the use of a long term bone marrow culture (Whitlock and Witte, 1982) were used in my studies. The first model system is derived from the study of Schwartz and coworkers (1986a). Using a murine long term bone marrow culture system, Schwartz et al. (1986a) were able to show that v-Ha-ras and v-myc have a synergistic effect in transforming murine pre-B cells. In their study, pre-B cell lines carrying both oncogenes were capable of causing lymphoma in syngeneic mice at high frequency and with a short latency, whereas pre—B cell lines carrying v-Ha-ras alone only gave rise to tumors occasionally and with a prolonged latency. The latter result indicates that v-Ha-ras is not sufficient to cause pre-B cell lymphomas and other secondary events are required to facilitate tumor formation. It is this finding that prompted us to search for these secondary events by the following approach. First, Whitlock-Wine high density bone marrow was infected with a Moloney murine leukemia virus (MoMuLV) based vector carrying a v-Ha-ras oncogene with MoMuLV as a helper virus. Usually pre-B cell cells with a single specific v-Ha-ras integration were established. This in vitro transformation step presumably facilitates multi-step tumorigenesis and provides a molecular marker for further studies. Secondly, these pre-B cell lines were subjected to tumor “y. e I 2 challenges, and they gave rise to tumors occasionally. This step provided the opportunity to accumulate other mutations which may cooperate with v-Ha-ras in vivo in tumorigenesis. The involvement of secondary events was examined by southern and northern hybridization analyses with various probes of known oncogenes and growth-related genes. Nearly thirty DNA probes (as listed in the Appendix B) of oncogenes, tumor suppressor genes, growth factor genes, or the flanking region of frequent viral integration sites were used to screen for gene rearrangements. The second model system involved the introduction of v-Ha-ras with or without other oncogenes or genes with oncogenic potential into Whitldck and Witte high density bone marrow culture. In this model system, the transforming effect of each gene was studied directly. This model system involves less labor than the first model, although it suffers the shortcoming of all in vitro transforming systems, the lack of interactions with the full range of in vivo cell types. It is worth mentioning that the transformation potential of any oncogene identified from the first model can be easily tested in this system. A pre-B cell growth factor, interleukin-7 (IL-7), was chosen to study. In the first chapter of this thesis, I will summarize briefly the methodologies that have been used to identify putative oncogenes that may be involved in tumorigenesis, and to test the oncogenic potential of these putative oncogenes. This is followed by a description of the known involvement of oncogenes and other molecular events in human B cell neoplasia as well as B cell tumors of other animals. 3 The results of studies on oncogene cooperativity between v-Ha-ras and other oncogenes in the in vivo and in vitro transformation of murine B cells will be presented in Chapters 2, 3, and 4 in this thesis. Chapter 2 details a c-myc activation found in the in vivo tumor progression of some of the B cell tumors obtained from the first model. This work has been published and will be presented as a manuscript. Data that were not shown in the manuscript because of space limitations will be included in the Appendix B. Chapter 3 describes the isolation and characterization of a viral integration flanking region common to tumor cell lines. That locus may encode a putative oncogene. This work involves a collaboration with Marge Strobel and Nancy Jenkins (NCI) who did the chromosomal mapping, and will be presented as a manuscript and submitted for publication at a later date. Chapter 4 reports the effects of lL-7 in cooperation with v-Ha-ras in transforming mouse pre-B cells with the second model system. It will be also presented as a manuscript and will soon be submitted to Molecular and Cellular Biology. In examining the transformed phenotypes of pre-B tumor cell lines, at least two independent tumor cell lines showed an interesting lineage switch phenomenon, in which macrophage specific characteristics were found. Since lineage infidelity has also been described in a minority of hematological malignancies (McCulloch, 1983), we decided to further characterize these two lines with the hope that some correlations can be drawn to the clinical cases, and to provide some insights on lineage determination in hematopoiesis. This work includes molecular, cytological, and functional analysis of the lineage switched HA w I 4 lines. I had made the discovery of the morphological changes of these tumor cell lines and subsequently subcloned one of these tumor cell lines, the T4 cell lines. I also proved that the T4 subclones were indeed descendants of their parental pre- 8 cell line, the R2 cell line, and did some of the cytochemical and immunocytological staining analyses on the T4 subclones. The demonstration of macrophage specific gene expression of T4 cells and kappa chain rearrangement of T4 subclones were also performed by me. Other portions of this work were contributed by the other authors listed on this manuscript. This work is in press in Developmental Immunology. I will present it as the submitted manuscript in Appendix A. At the end, a section of summary and discussion has been included to cover points that can be integrated from all of the above work but were not specified in each individual chapter. Reference: McCulloch EA. (1983). Stem cells in normal and leukemic hemopoeisis. Blood 62, 1-13. Schwartz R.C., L.W. Stanton, S.C. Riley, K.B. Marcu, and ON. Witte. (1986a). Synergism of v-myc and v-Ha-ras in the in vitro neoplastic progression of murine lymphoid cells. Mol. Cell. Biol. 6, 3221-3231. Whitlock C.A., O.N. Witte. (1982). Long-term culture of B lymphocytes and their precursors from murine bone marrow. Proc. Natl. Acad. Sci. USA 79, 3608-3612. Chapter 1 Literature review In this chapter, evidence in support of the multi-step nature of tumorigenesis will be described first. It is followed by a brief overview of methodologies used in the identification of oncogenes and the evaluation of their oncogenic potential. Comparison of the use of in vivo versus in vitro model systems, and animal versus human systems is included in this section. Then, I will discuss in detail the literature on oncogenes known to be involved in the formation of B lymphoid tumors. The discussion will encompass oncogene activation observed in tumors of human and other animals, as well as in in vitro transformation of B cells. 1. Tumorigenesis Is a multiple-step process Cancer cells differ from their normal counterparts in that they no longer respond to normal growth controlling mechanisms. Since the proliferation and differentiation of somatic cells in higher organisms are regulated by multiple controls, cancer cells probably are the end result of multiple changes which may take years to develop. Several lines of evidence confirmed that the natural history of spontaneously occurring human and animal cancers is usually a multi-step process. First, statistical analyses have shown a rapid increase in the incidence of cancer with age among most of the important human cancers (Fisher and Holloman, 1951; Armitage and Doll, 1954; Dix, 1989). Results of these analyses suggest that multiple events may accumulate with time, and thus are responsible for the increased cancer rates in old age. Peto et al., (1975) further demonstrated 9) kn ”u. .. ”he 6 that the intrinsic effects of ageing such as falling immunological surveillance or age- related hormonal changes were not required to explain the vastly increased incidence of cancer in old age, and that age was a reflection of the duration of exposure to carcinogenic agents. They used mice at 10, 25, 40, and 55 weeks of age in a skin carcinogenesis experiment with benzpyrene. The rate of malignant epithelial tumors in each group increased steeply with time and the increase was independent of age at the start of exposure. Second, studies of chemical carcinogenesis have defined stages into which carcinogenesis may be divided: initiation, promotion and progression (Diamond at al., 1980; Pitot, 1990). Each stage is induced independently by a different class of chemical agents. Third, histological evidence can somewhat arbitrarily be used to divide the neoplastic development into three phases: initiating, intermediate, and advanwd (Foulds, 1975). The initiating phase is either clinically "silent" or manifested only by apparently trivial and dubiously neoplastic lesions such as hyperplasia. The intermediate stage is characterized by the emergence of "precancerous" or "premalignant" lesions which may progress into the advanced stage, or persist indolently for a long time with minimal growth and no qualitative change, or regress completely. The last stage is characterized by the presence of malignant carcinomas or sarcomas. The phenomenon of tumor regression suggests the requirement of multiple factors for preneoplastic cells to develop a fully malignant phenotype. Fourth, cytogenetic studies of tumors reveal a multiplicity of chromosomal abnormalities in all human cancers (Mitelman, 1991; Solomon et al., 1991). r- JP :60! U test; ("I 7 Examination of colorectal carcinoma (Fearon and Vogelstein, 1990) and neuroblastoma (Knudson and Meadows, 1980) indicate that additional cytogenetic lesions are associated with the advanced stage of these malignancies. Finally, the most direct evidence comes from the discovery of chicken RNA tumor viruses, AMV-E26, MH2, and AEV-ES4 strains, which carry dual oncogenes (ets-myb, myc-mil, and erb-A-erb—B, respectively). The full oncogenic potential of these viruses requires both oncogenes (Cole et al., 1983 and Jansen et al., 1983 for MHZ; Leprince et al., 1983, Nunn et al., 1983 and Kan et al., 1983 for E26; Frykberg et al., 1983 for ES-4). Viruses possessing only one of these oncogenes shows more limited tissue specificity and longer latency for tumor development. Similar oncogene cooperation in tumor development has also been demonstrated in DNA transfection experiments. Land and coworkers (1983a; 1983b) showed that myc and ras could fully transform primary rat embryo fibroblasts when transfected together but not singly. Schwartz et al. (1986a) further extended the synergistic effect of the myc and ras oncogene to the transformation of murine B cells. Other examples of oncogene cooperation involved in tumorigenesis have recently been summarized by Hunter (1991). In conclusion, through decades of studies, researchers have been able to correlate the multifactorial characteristics of neoplasia from the histological and cellular levels to the molecular level. Identification of genetic lesions and their effects in tumorigenesis not only benefits our understanding of the growth and differentiation of normal and cancerous cells, but also provides insights on prevention or therapy of cancer. 8 2.1 The search for genes that are involved in tumorigenesis Studies of mechanisms of chemical carcinogenesis (Pitot, 1990; Balmain and Brown, 1988) and heritability of cancers (Ponder, 1990) have strongly suggested that the lesions of tumors reside in the genetic material. The search for putative oncogenes in a genome size of 3 x 109 base pair containing approximately 10‘ to 105 genes was greatly simplified by the discovery of viral oncogenes in animal RNA tumor viruses. The first RNA tumor virus was isolated by Rous (1910), but it was not until the development of molecular biology that the identification of the v-src oncogene became possible (Stehelin et al., 1976a). Twenty six viral oncogenes have been identified from animal RNA tumor viruses (Varmus, 1989), and shown to exhibit sequence similarity with cellular genes (later termed protooncogenes) by hybridization and sequencing analyses. Since the cellular protooncogenes are also found to be conserved among different species, it stresses the important roles that these genes may play in regulating growth and differentiation. Thus, alteration or destruction of these genes may contribute to tumor progression. The list of protooncogenes has been expanded by four major approaches (i) gene transfer, (ii) analysis of known chromosomal translocations or amplifications, (iii) mapping of viral integration sites in virally induced tumors, (Iv) sequence homology to the known oncogenes. Some of these genes were repeatedly identified by different methods. 2.1.1 Identificatlon of cellular oncogenes by DNA transfer experiments The most common method of gene transfer has utilized DNA transfection 9 and focus formation (Graham and van der Eb, 1973; Shih et al., 1979). DNA is isolated from tumor cells and introduced into recipient cells, usually an immortalized mouse fibroblast line, NIH3T3. NIH3T3 cells have two major advantages other than their easy cultivation: (i) they are flat, contact-inhibited cells that form a monolayer culture, and (ii) their DNA can be distinguished in the hybridization experiments from the DNAs of other species, thus allowing identification of donor DNA. The first property allows one to examine the transfected cultures for the appearance of morphologically altered (transformed) foci due to the expression of an introduced oncogene. Examples of cellular oncogenes identified by this procedure are N-ras from neuroblastoma (Shimizu et al., 1983), trk from colon carcinoma (Martin-Zanaca et al., 1986), and others as listed in Table 1.1. Oncogenes of the ras family predominate among those found fin about 20% of human tumors tested)(Der et al., 1982; Parada et al., 1982; Santos et al., 1982), whereas cellular analogues of other viral oncogenes are less frequently detected by this method. This could be due, at least in part, to the ability of NIH3T3 cells to respond morphologically to a given oncogene product, to the need for the gene to be genetically dominant, and to the need for the gene to be sufficient for the expression and the maintenance of the transformed state. In other words, NIH3T3 cells may be the wrong lineage or species to respond to the effects of certain oncogenes. In addition, NIH3T3 cells may have a preneoplastic phenotype as they already have the ability to grow continuously in culture. Thus, they may be more susceptible to genes whose effects are manifest during the later stages of tumor progression. 10 Table 1.1 Oncogenes identified by DNA transfer experiment Gene Sourcec Function Reference N-ras Neuroblastoma G-protein-like Shimizu et al., 1983 neu Rat neuroglloblastoma EGF receptor like Shih et al., 1981 Bargmann et al., 1986 mas Epidermoid carcinoma Angiotensin receptor Young et al., 1986 hst Kaposi’s sarcoma FGF family member Delli Bovi et al., 1987 Taira et al., 1987 trk Colon carcinoma Receptor-like Martin-Zanaca at al., 1986 met Osteosarcoma cell line Receptor-like Cooper et al., 1984a ret T cell lymphoma Receptor-like Takahashi et al.. 1985 dbla Diffuse B cell lymphoma Cytoskeletal matrix Eva and Aaronson. 1985 associated phosphoprotein Graziani et al., 1989 mcf.2a Mammary carcinoma cell line as that of dbl, a Fasano at al., 1984 Ice Hepatocellular carcinoma b Ochiya et al., 1986 mel Melanoma cell line b Padua at al., 1984 raf Stomach cancer Serine/threonine kinase Shimizu et al.. 1985 res Mammary carcinoma cell line Receptor-like Birchmeier at al.. 1985 a: these two genes were found to be two different activated versions of the same protooncogene (Noguchl et al., 1988) b: not yet defined c: human tumors if not specified A 2.1.2 I 11 In another aspect, oncogenes that are isolated from RNA tumor viruses carrying a single oncogene probably contain multiple mutations within each oncogene and, therefore, are capable of inducing tumors as a single gene. Thus, the failure of detecting cellular counterparts of other viral oncogenes by this method may simply be attributed to the fact that the cellular counterparts did not contain multiple mutations to induce focus formation of NIH3T3 cells. 2.1.2 Characterization of viral flanking regions Studies of RNA tumor viruses such as Avian leukosis virus (ALV), Moloney murine leukemia virus (MoMuLV), and Feline leukemia virus (FLV) that do not carry oncogenes reveal another mode of transformation. Insertion of the viral genome into a cellular genome may activate adjacent cellular genes through enhancer or promoter sequences of the viral LTR (Hayward et al., 1981; Fung et al., 1981; for a review see Nusse, 1986a). Therefore, cloning of flanking regions of frequent viral integration sites may allow the identification of putative oncogenes. A variety of genes and “loci" have been isolated by this method. Protooncogenes which contain homology to known viral oncogenes and other known cellular genes are shown in Table 1.2, and "loci" that do not show homology to known viral oncogenes and other known cellular genes are shown in Table 1.3. These “loci" may correspond to novel oncogenes or may be linked to protooncogenes at a distance. Among the "loci" listed in Table 1.3, int-1, int-2, and pim-1 have been shown to contain detectable transcriptional activity and to encode proteins with distinct functions (Nusse et al., 1984; Dickson et al., 1984; Cuypers et al., 1984; Selten et Tabl J «n- p v I? Table 1.2 Cellular genes activated by proviral Insertion 12 Gene Source Virus Reference c-myc Chicken bursal lymphoma ALV Hayward et al., 19813 Mouse T cell lymphoma Souls-MuLV Adams et al., 1982‘ Cat T cell lymphoma FeLV Nell et al., 1984a N-myc Mouse T cell lymphoma Moloney MuLV van Lohuizen et al., 1989a c-erbB Chicken erythroleukemia ALV Fung et al., 1983 c~myb Mouse lymphosarcoma defective MoMuLV Shen-Ong et al., 1984 c-Kl—ras Mouse myeloid cell line Friend MuLV George et al., 1986 c-Ha-ras Mouse T cell leukemia Moloney MuLV Ihle et al., 1989 lL2 Ape T cell lymphoma cell line GaLV Chen et al., 1985 lL-3 Mouse myelomonocytlc leukemia IAP Ymer et al., 1985 CSF-1 Mouse monocyte tumor endogenous Baumbach et al., 1988 ecotropic provirus GM-CSF Mouse promyelocytic cell line lAP,R-MuLV,F-SFFV Stocking et al., 1988 c-mos Mouse plasmacytoma line IAP Canaanl et al., 1983 p53 Mouse erythroleukemlc cell line Friend MuLV Hicks and Mowat, 1988 abbreviation: MuLV: murine leukemia virus IAP: intracisternal A particle FeLV: Feline leukemia Virus ALV: Avian Leukosis Virus R-MuLV: Rauscher murine leukemia virus F-SFFV: Friend spleen focus forming virus GaLV: Gibbon Leukemia Virus ‘2 only the earliest reference is shown T: .K e ‘ s A. a C- A\.V 13 Table 1.3: VIraI flanking regions without viral oncogene analogues. Name Source Virus mRNA Reference Evi-I AKXD myeloid tumor Cas—Br-M MuLV Mucenskl et al.,1988a MCF yes Morlshita et al., 1988 MM NIH/Swiss or SIMS Moloney MuLV nd Poirier et al., 1988 pre-B cell lymphoma Fis-f AKR lymphoma Friend MuLV nd Silver and Kozak, 1986 ' or BXD-2 leukemia Dsi-f Fisher rat thymoma Moloney MuLV nd Vijaya et al., 1987 Pim-2 Balb/c or Moloney MuLV nd Breuer et al., 1989a CS7BL10 lymphoma Pim-I Balb/c or AKR T lymphoma MCF yes Cuypers et al., 1984 Int-1 03H Mammary carcinoma MMTV yes Nusse and Vannus, 1982 Int-2 C3H Mammary carcinoma MMTV yes Dickson et al., 1984 Int-41 mouse mammary and MMTV yes Garcia et al., 1986 kidney adenocarcinoma MIvi-t' rat thymoma Moloney MuLV nd Tsichlls et al., 1983a (PW-1) MIvi-2 rat thymoma Moloney MuLV nd Tsichlls et al., 1984 MIvl-3 rat thymoma Moloney MuLV nd Tsichlls et al., 1985a MIvi-4 rat thymoma Moloney MuLV nd Lazo et al., 1990 Gin-1 mouse thymoma Moloney MuLV nd Villemur et al., 1987 Spi-t mouse erythroleukemia SFFV yes Moreau-Gachelln at al.,1988 Bic Avain B cell lymphoma ALV yes Clunnan and Hayward, 1989 Abbreviation: MuLV and ALV: same as those in Table 1.2 MMTV: Mouse mammary tumor virus MCF: mink cell focus forming virus Cas-Br-M MuLV: Casitas Brain Mousetropic MuLV SFFV: spleen focus forming virus ‘: also known as mis-f 14 al., 1986; Nusse, 1988), and to exhibit transforming ability in transgenic mice (Adams and Cory, 1991) or the in vitro transformation of an epithelial cell line (Int-1 only; Brown et al., 1986). On the other hand, many of them do not have detectable transcriptional activity. Since the enhancer element of the viral LTR may function over a long distance, it is possible that certain putative oncogenes may be located distal to the breakpoints of viral integration sites. Techniques such as chromosomal walking, jumping and YAC cloning may facilitate the identification of such genes. Additionally, exon trapping (Duyk et al., 1990; Buckler et al., 1991), a method used to identify any exon sequence from cloned genomic DNA may also be used to facilitate the identification of any transcription unit. Once a transcription activity is associated with a “locus, it is, however, necessary to test for its transformation potential. Either the in vitro focus formation assay (see DNA transfer experiment) or the transgenic animal model (see below) can be used for that purpose. The spectrum of oncogenes that can be identified by this method relies on the randomness of viral integration. Although retrovirus integration has been shown to have preferred target sites (Shih et al., 1988), these preferred sites comprise only 20% of all the viral integration sites. Thus, this preference does not seriously affect the variety of oncogenes that may be identified by this method. In addition, the nature of this preference is unknown. Perhaps it resides in the active transcription of preferred regions. The fact that both cellular counterparts of viral oncogenes and novel oncogenes are isolated by this method makes it a feasible way of searching for a diverse class of genes that are involved in the regulation of growth and differentiation. It has clearly allowed identification of a wider variety of 3‘1 or 15 oncogenes than transfection and focus formation experiments. Interestingly, no human oncogene identified thus far is activated or inactivated by viral integration. However, the discovery of multiple copies of endogenous virus-related sequences in human cells suggests that gene activation through this mode may still be possible (Bonner et al., 1982; Callahan et al., 1985). 2.1.3 Molecular Characterization of chromosomal aberrations Chromosomal aberrations have long been associated with human cancers. Karyotype analyses of metaphase chromosomes has found some abnormalities specific for distinct tumor types (for a recent review, Mitelman, 1991). The discovery of the Philadelphia (Ph1) chromosome in bone marrow cells of patients with chronic myelogenous leukemia (CML) actually laid the foundation for the association of specific chromosomal anomalies with a particular neoplasm (Nowell and Hungerford, 1960). It was not until recently, however, that the molecular basis of these cytogenetic lesions has been elucidated as a result of the improvement of both cytogenetic and molecular cloning methods. Methotrexate treatment (aminopterin) (Yunis, 1976; Hagemeijer et al., 1979) is to block DNA synthesis pathways is blocked so that cells can be collected at the S phase of a cell cycle. The block is then released by adding thymidine to allow DNA synthesis using the salvage pathway. A large number of cells enter mitosis synchronously following these treatments. These synchronized cells are then treated briefly with colchicine followed by standard banding procedures (Chaudhuri et al., 1971; Caspersson et al., 1970; Hsu, 1974). Chromosomes obtained by this procedure are less condensed, allowing 1200 bands to be identified. The increased banding IVA Ni V t:- Va; 16 resolution not only increases the accuracy of identification of chromosomes, but also leads to the recognition of many karyotypic changes and the association of these changes with particular neoplasias. Cloning of genes located in translocation junctions or other chromosomal aberrations has been done by conventional cloning methods, ie. use of probes for genetic markers near the locus of interest, and chromosomal walking using a genomic library until the authentic gene is identified by in situ hybridizations or southern blot analyses. The introduction of microdissection and microcloning should advance this process. Microdissection was first used by Scalenghe et al.(1981) to clone DNA from Drosophila polytene chromosomes. It was later used in the human genome mapping project (Bates et al., 1986; Kaiser et al., 1987), and to clone genes involved in human genetic diseases (LOdecke et al., 1989; Kondo et al., 1984). In this technique, a very small segment of the chromosome of interest can be dissected with needles from banded chromosomes by using an electronically controlled micromanipulator. Use of this method in combination with microcloning in which the technique of polymerase chain reaction (PCR) was incorporated (Edstrom et al., 1986) can reduce the number of genomic clones required to screen for the locus of interest compared to that in conventional cloning procedures. Adapting these approaches may greatly facilitate the cloning of genes that reside at the site of chromosomal lesions. A recent review by Solomon et al. (1991) summarizes genes identified from chromosomal aberration sites (Table 1.4). Many of them are either analogues of viral oncogenes, or genes involved in the cell cycle or differentiation. More Tab (5 ) (b -3 1 I I!" " a... c A Axe“: v. _ l L‘ . 1 “1 Ta' : L .U A“. IU- ‘: N’tnd U 4 F. ,tf U l 33:4 17 Table 1.4 Genes Identified from chromosomal aberrations Gene Source Protein type Reference chromosomal amplification N-myc L-myc Neuroblastoma Small-cell lung cancer Nuclear protein Nuclear protein Schwab et al., 1983 Little et al., 1983 chromosomal translocation or inversion (adapted from Table 1 In Solomon et al. (1991)) Gene Disease Rearrangement Protein type myc Burkitt lymphoma t(8;14)(q24;q32) HLH domain t(2;8)(pi 1 ;q24) t(8;22)(<124;q11) T-ALL t(8;14)(q24;q11) BcI—I B-CLL t(11;14)(q13;q32) GI cyclln-Ilke Bel-2 Follicular lymphoma t(14;18)(q32;q21) Inner mitochondria membrane BcI-3 BCLL t(14;19)(q32;q13) CDC 10 motif lL-3 Pre-B ALL t(5;14)(q31;q32) Growth factor Lylt T-ALL t(7;19)(q35;p13) HLH domain TcI-s T-AII t(1 ;14) (p32) (q1 1) HLH domain Rbntl T-ALL t(11;14)(p15;q11) UM domain Rbnt2 T-ALL t(11;14)(p13;q11) UM domain Tan1 T-ALL t(7;9)(q35;q34) Notch homolog Hox11 T-ALL t(10;14)(q24;q11) Homeo domain Pth Parathyroid adenoma inv(11)(p15;q12) Deregulate myc Big-1 B-CLL t(8;12)(q24;q22) Deregulate myc Bcr-Abl CML, B-ALL t(9;22)(q34;q11) Bcr. Gap for p21'“ Abl, Tyrosine kinase Pml-Rara APL t(15;17)(q22;q11-12) Pml, Zinc finger Rare, Zinc finger Dek-Can AML-M2,-M4 t(6;9)(p23;q34) Dek, nuclear protein Can, cytoplasmic protein EZA-Pbx Pre-B ALL t(1;19)(q23;p13) 52A, HLH domain Pbx, homeodomain Rel-Nrg NHL lns(2;2)(p13;p11-I4) Rel, NF-kB family Nrg, no homology Abbreviation: CLL: chronic lymphocytic leukemia ALL: acute lymphocytic leukemia CML: chronic myelogenous leukemia AML: acute myelogenous leukemia NHL: non Hodgkin’s lymphoma {1‘0 " at": i839 MJ‘: U‘v 2.1. (L) r I) I..." U 18 significantly, tumor suppressor genes (such as RB and WT-I; Marshall, 1991, recently reviewed by Weinberg, 1991) and genes associated with apoptosis (such as bcI-2) were also discovered. Alteration of the genomic structure of these genes may deregulate their expression and participate in the transformation of cells, and consequently lead to tumor formation. 2.1.4 Identification of new oncogenes by homologous sequences Another approach not commonly used, but potentially useful, is to search for sequences homologous to known oncogenes assuming the conservation of important regulatory genes. Given the fact that all the genes in the src family (Hunter and Cooper, 1985) and ras family (Barbacid, 1987) share significant homologies among certain genetic domains (such as the effector domain of ras genes and the carboxy-terminal region of src-related genes), hybridization probes for those regions may identify new members of these gene families. The Ick oncogene (Marth et al., 1985) was first isolated from a murine T cell lymphoma by using an oligonucleotide probe for the conserved major tyrosine phosphorylation site. Other proliferation and differentiation related genes may also be identified from cells on the basis of reversion from the transformed phenotype. Such cells have been generated either by cell-fusion with normal cells (Harris, 1988) or DNA transfection. An example of a gene identified by the latter method is the Krev-I gene, which is responsible for the revertant phenotype of Kirsten sarcoma virus- transformed NIH3T3 cells (Kitayama et al., 1989; Noda et al., 1989). The oncogenic potential of the putative oncogenes identified from the above 19 procedures should be tested to verify their active roles in oncogenesis. The following sections describe general approaches used for this purpose. 2.2 Experimental models for transformation and tumor progression 2.2.1 Use of animal versus human models Animal model systems are obviously required for experimental in vivo carcinogenesis studies, since it is ethically unacceptable to use human subjects. The compatibility of results from animal models with those obtained in humans has been supported by several observations. First, the conservation of oncogenes among species suggests their common properties and regulation (Varmus, 1989). Additionally, human oncogenes are able to transform mouse fibroblasts in DNA transfection experiments (see above) and induce tumors in transgenic mice (Palmiter and Brinster, 1986). Second, growth factors and growth regulatory mechanisms do not appear to be species specific since transplanted tumors from a great range of species can grow in immunodeficient, athymic (nude) mice while normal tissue can not (Stiles and Kawahara, 1978). These results suggest the universality of the genetic events causing neoplasia, and the validity of animal models in studying human carcinogenesis. The fact that laboratory animals can be environmentally and genetically controlled makes them an even better system to work with. On the other hand, the use of human tissues and cells does offer some unique advantages. First, some rare forms of human cancer reflect inherited, predisposing conditions. Their genetic basis and perhaps common pathways to carcinogenesis may be understood through the study of nontumorous cells from ANN K... 20 affected individuals (Ponder, 1990). Moreover, the fact that human cells are genetically more stable in vitro than most rodent cells (DiPaolo, 1983) makes them especially suitable for studying the multiple steps of tumorigenesis. Lastly. the findings from studies of human cells complement and validate the results derived from studies of laboratory animals. 2.2.2 Use of in vivo versus in vitro systems The role of the host, not only in controlling, but also in facilitating the development and spread of cancer has already been established (for a review, see Alexander, 1987). These host factors interact with the genetic lesions that reside in the tumor cells themselves. The in vivo system therefore has an essential role in cancer studies because the integral multi-systemic interactions of the organism remain intact. The disadvantage of using in vivo models for studies of tumor progression is its inconvenience for dissecting events within the progression. Most often an endpoint must be chosen for analysis and intervening events must be implied. An in vitro model using cell or tissue culture, however, provides opportunities to dissect the processes controlling growth, differentiation, neoplastic transformation and tumor progression of cells, and to describe their mechanisms in biochemical or molecular terms. 2.2.3 Model systems used to test the oncogenic potential of oncogenes One commonly used in vitro culture system, NIH3T3 cells, although allowing the detection of certain oncogenes, presents limited sensitivity since this method tends to identify oncogenes in the ras gene family as discussed previously. LEG.” AT; 4 3"" ' U A~F ‘ v 't‘ J 21 Various tissue culture systems including those of epithelial and hematopoietic origins (Gabrielson and Harris, 1985; Rheinwald and Green, 1975; Deeh, 1985; Lechner “T21%* ’eck, 1985; Dexter et al., 1977; Whitlock and Witte, 1982) were established and have allowed the study of gene interactions within differentiated cell types. Among them, the long-term B cell culture system of Whitlock and Witte (1982) is exclusively used in our studies. This culture system can provide not only a wide range of targets for transformation within the B cell developmental series, but also allows the culture of cells that are not transformed or of an intermediate transformed phenotype. It thus provides an excellent tool to test the transforming potential of oncogenes in the B cell lineage, and to elucidate the mechanisms of transformation on the molecular level. In vitro human B cell lines such as the lymphoblastoid cell lines (LCLs) are commonly used for the same purpose. These cell lines are derived from the infection of human peripheral blood with Epstein-Barr virus (EBV) (for review see Nilsson and Klein, 1982). Examples of the uses of these lines in testing oncogenic potential and oncogene cooperativity of putative B cell oncogenes are described in human B cell neoplasia (see below). Transgenic mice can also be used to assess the transforming potential of various oncogenes (for review see Palmiter and Brinster, 1986; Adams and Cory, 1991). Using tissue specific regulatory elements, one can examine the oncogenic potential of the gene of choice in a particular tissue. For example, the enhancer sequences of immunoglobulin genes (Eu) have been used to test oncogenes such as c-myc and bcI-2 in the oncogenesis of B-cell neoplasia (Adams et al., 1985; 22 McDonnell et al., 1989). In addition to its advantages as an In vivo system (as described previously), it also provides the only means for studies of tissue specific oncogenesis where an in vitro culture system is lacking. 2.2.4 Model systems used to study tumor progression Two of the most established genetic systems, the mouse skin carcinoma (for a review see Balmain and Brown, 1988) and human colon carcinoma (Fearon and Vogelstein, 1990), share the advantage of ease in access to and recognition of tumor samples at different stages of progression. In contrast, tumors of lymphoid and other organs lack a good animal model to study molecular changes during the tumor progression period, mainly due to the difficulty of identifying preneoplastic cells and obtaining cells at varying stages of neoplastic development in a single host (i.e., the collection of neoplastic samples of these tissue types often requires termination of the donor animal). Even if the experimental animals were blindly sacrificed at different time points to collect tumors at different stages of neoplastic development, one still encounters the problem of insufficient numbers of cells to perform molecular analysis. In vitro cultures would be required to expand these isolates and such in vitro tissue culture systems are lacking for many of these tissue types. Although the results generated from the above in vivo systems may be applicable to tumors of other origins, evidence of the association of certain oncogenes with tumors of particular tissue types from studies of transgenic mice and inherited cancers (Adams and Cory, 1991; Marshall, 1991; Lanes et al., 1981 & 1982; Cooper and Neiman, 1980; Padhy et al., 1982) have suggested the need for tissue specific model systems. Although the use of 23 transgenic mice has proven to be a useful in vivo model to test the ability of an oncogene to initiate tumor formation, little knowledge is gained about the secondary events that have contributed to the progression of tumors. Tests of oncogene cooperativity either involve the construction of transgenic mice with multiple oncogenes or performing crosses with transgenic mice carrying different oncogenes (Adams and Cory, 1991). These procedures may involve intensive labor. Moreover, the cooperative effect is unavoidably under the influence of unnatural environments because cells of all tissue types may express the activated oncogenes. The use of tissue-specific enhancer sequences may avoid this problem; however, these sequences are not available for all tissue types. One approach used to study oncogene cooperation in the tumor progression of transgenic mice is to infect transgenic mice with retroviruses to facilitate tumor progression (van Lohuizen et al., 1991). The Whitlock and Witte culture system (1982) allows certain advantages in the study of oncogene cooperativity. By comparing the transformed phenotypes of B cells generated after the introduction of two or more oncogenes of interest into this culture to those of B cells generated from cultures with a single oncogene, one can determine whether one oncogene can complement another in transforming B cells. This procedure is relatively easy as compared to that of transgenic mice. It also provides a faster way of testing whether there are temporal effects on the acquisition of genetic alterations in tumor progression. In summary, using the methodologies described above, extensive studies have been performed to uncover the basis of tumorigenesis. Among them, hematopoietic neoplasias are the best characterized tumors at the molecular WE 24 genetic level due to their frequent occurrence in animal models and their high mitotic index, allowing for cytogenetic analyses (see above). Many alterations of protooncogenes have been specifically associated with different subtypes of leukemias/lymphomas of different species, and are summarized elsewhere (Schwartz and Witte, 1988; Solomon et al., 1991; Sawyers et al.,1991). Here, I will describe exclusively the molecular characterization of B cell lymphomas/leukemias of human, mouse, and avian origin, since this has been the focus of my thesis. 3. Multiple genes or factors that are Involved in B cell lymphomas/leukemias of human, mouse, and avian 3.1 Human B cell neoplasia 3.1.1 Burkitt’s lymphoma Burkitt’s lymphoma (BL) is the best characterized human B cell tumor. It was first described by Burkitt (1958) as a distinct clinicopathological entity occurring with high frequency in the jaws of children from Central Africa. Non- endemic (or sporadic) BL is also found in areas outside of Africa but the rate of incidence is low (O’Conor et al., 1985; Philip et al., 1982). BL is a malignant lymphoma comprising a monomorphic outgrowth of B lymphocytes. ‘lhe stage of maturation of BL cells varies among individual tumors, from a near pre-B phenotype (Preud’homme et al., 1975) to a more mature phenotype with expression of predominantly lgM (Klein et al., 1967), and occasionally IgD, IgG, or even IgA (Preud’homme et al., 1985). However, the cells never fully differentiate to a plasmacytic phenotype. The etiology of BL involves at least two steps. Infection with Epstein-Barr Virus (EBV) appears to be the primary event in the development of African Burkitt’s (TI .E=l VII 25 lymphoma from both genetic (Geser et al., 1983) and epidemiological evidence (de-The’ et al., 1978; Geser et al., 1982). Geser and coworkers found that 96% of African Burkitt’s lymphomas carry multiple cepies of the EBV genome in all their cells. Both groups showed that a high multiplicity EBV infection early in the life of African children contributes to the tumor development. Raab—Traub and Flynn (1986) and Neri et al. (1991) have studied the termini of EBV episome genomes in both endemic BL primary tumors and cell lines. The linear EBV DNA has variable numbers of direct tandem 500 bp repeats at each terminus. Restriction endonuclease analyses have indicated that the termini are uniformly clonal in all cases. This result again strongly suggests that EBV infection has preceded, and thus, most likely contributed to clonal expansion in these malignancies. Although the lack of viral markers in 4% of the endemic BL and approximately 85% of the non-endemic BL cases (Lenoir et al., 19843) may argue the against a necessary causal relationship between EBV and BL, the fact that EBV can transform human peripheral B cells into established cell lines (T akada and Osato, 1979; Robinson and Smith, 1981) and that EBV can induce B cell lymphomas in marmosets (zur Hausen, 1980) implies an important role for EBV in BL. The immortalizing mechanism of EBV has been suggested to be mediated through autocrine stimulation by the finding of the release of a Bee" growth factor (BCGF) from Burkitt lymphoma cells and EBV infected cell lines (Gordon et al., 1984). The precise consequences of this autocrine stimulation are unknown. However, two EBV-encoded proteins are found to be involved in the process of oncogenesis. The EBNA2 gene product, a nuclear protein, is required for immortalizing B cells since EBV deleted for this gene fails to do so (Menezes et al., 26 1975; Delius and Bornkamm, 1978; Bomkamm et al., 1980; Heller et al., 1981). EBNA2 can also transiently stimulate cellular DNA synthesis upon transfection into B cells (Volsky et al., 1984). Another EBV encoded protein, LMP1 (a latent membrane protein), can transform established rodent cells (Wang et al., 1985; Baichwald and Sugden, 1988). The linkage between the release of the BCGF and the biochemical effects of these two proteins are becoming clear through the following studies. EBNA2 is a multiple-function protein, and may act similarly to T antigen of SV40 virus. EBNA2 was found to induce CD21, CD23, and LMP1 expression upon introducing this gene into cell lines which are infected with EBV strains, deletion mutants that do not encode EBNA2 protein (Wang et al., 1990; Abott et al., 1990). CD21 is a surface protein involved in human B cell differentiation (T edder et al., 1984). CD23 is a membrane-bound B cell activation marker, and acts as an autocrine BCGF for normal and transformed B cells when shed (Swendeman and Thorley-Lawson, 1987). LMP1 was found to cooperatively Induce 0023 expression, and to induce the expression of several cellular adhesion molecules (Wang et al., 1990). The increased expression of the cellular adhesion molecules may explain the phenotypic changes of LMP1 transfected cells including growth in large tight cell clumps (Wang et al., 1988). Taken together, these findings suggest that EBNA2 may immortalize B lymphoid cells by a autocrine mechanism through induction of CD23. This autocrine stimulation is augmented through the cooperation with LMP1, which may act via cellular second messengers. Whether EBNA2 acts directly or through other cellular intermediates in the upregulation of CD23 transcription remains to be determined. The second step in the development of Burkitt’s lymphoma may involve ' A; fls'y\ U“ '. MFA I‘y’Y‘. 7' F x e": Crag 27 deregulation of the c-myc gene through chromosomal translocations of chromosome 8 on which it residues. The almost universal presence of chromosomal translocations between chromosome 8 and one of three other chromosomes (chromosome 14, 2 and 22) is another characteristic of Burkitt lymphoma. Only one exception to this has been reported (Zech et al., 1976). Translocation t(8;14)(q24;q32) was found in approximately 80% of cases of Burkitt lymphoma (Croce and Nowell, 1986), while the remainder of these tumors carry t(2;8)(p11;q24) or t(8;22)(q24;q11) translocations. It is striking that each of these translocations involves the cytogenetic location of one of the immunoglobulin (lg) loci. The heavy-chain gene is located at chromosome 14q32 (Croce et al., 1979); the kappa and the lambda genes are at 2p11 (Malcolm et al., 1982) and 22q11 (Erikson et al., 1981), respectively. This may suggest a molecular relationship between the rearrangement of Ig genes and oncogenesis, perhaps through a aberrant recombination between two chromosomes. The identification of the crossover point of the translocation on chromosome 8 was facilitated by the discovery in murine plasmacytomas of a similar translocations (see below). The involvement of the c-myc gene in these chromosomal translocations was suggested by the finding of c-myc gene activation in an abortive immunoglobulin gene recombination in a mouse plasmacytoma (Shen-Ong et al., 1982). Additionally, the c-myc gene was mapped to chromosome 8q24 using human/rodent somatic cell hybrids by Dalla Favera et al. (1982). Finally, southern blot analyses showed that the c—myc gene was rearranged in approximately 50% of Burkitt’s lymphomas examined (Bernard et al., 1983; Dalia Favera et al., 1983; Taub et al., 1982). Molecular cloning ultimately enabled the detailed analysis of the 28 translocation break points and demonstrated that the translocations join sequences from the lg genes to regions surrounding c-myc (reviewed by Haluska et al., 1987). The result of all three translocations is the constitutive expression of the c- myc gene. In contrast to the high levels of c-myc expression associated with the viral transformation of avian B-cell lymphomas (see below) and human tumor cell lines that contain amplified c-myc genes (Collins and Groudine, 1982; Delia Favera et al., 1982), high levels of c-myc expression are not found consistently in all Burkitt’s lymphoma cell lines. A variety of mechanisms have been proposed to account for the myc/lg juxtaposition in its contribution to tumorigenesis (review by Klein and Klein, 1985). These include abnormally high transcription rates, abnormal transcription size, changed promoter usage, mutations, and lack of transcriptional pausing at exon 1 of c-myc gene. None of the above mechanisms seems to be applicable universally to all or even most of the tumors. The current idea is that c-myc is deregulated as a consequence of cis-acting sequences associated with the constitutively active lg region. The result of such deregulation would be to keep the cells in continuous division through a loss of c—myc’s normal regulation. The oncogenic potential of c-myc genes in Burkitt’s lymphoma was tested by Lombardi et al. (1987). They introduced an expression vector containing constitutively expressed c-myc into EBV-infected human B lymphoblastoid cell lines (LCLs)(Nilsson and Klein, 1982). The resulting myc-transfeCted LCLs displayed a reduced serum requirement for growth, an increased soft-agar cloning efficiency, and an increased tumorigenicity in nude mice as compared to those of vector-only- 29 transfected LCLs. Their results indicate a contribution of the c-myc gene in the tumorigenesis of Burkitt’s lymphoma, and a cooperation between two putative immortalizing functions. However, the inability of myc-transfected LCLs to form colonies as efficiently as BL cell lines, and their low tumorigenicity have suggested that additional events are required for a fully tumorigenic phenotype. In addition to the common events of EBV infection and c-myc translocation, p53 mutations were found associated with 9/27 biopsies of BLs and 17/27 BL cell lines (Gaidano et al., 1991). The p53 gene encodes a 53-kDa nuclear phosphoprotein that may be involved in the negative regulation of cell growth (Lamb and Crawford, 1986; for review see Iman and Harris, 1991). Additionally, p53 may differ from prototypical tumor suppressor genes in that at least some p53 mutant alleles can behave as dominant oncogenes by transforming target cells in vitro and causing tumorigenesis in transgenic mice even in the presence of the normal allele (Halevy et al., 1990; Lavigueur et al., 1989). Gaidano and coworkers’ results suggest both loss of function and dominant negative mutations are present in BLs.N-ras activation was found in a sporadic Burkitt’s lymphomas (Murray et al., 1983; Lenoir et al., 1984b). The effect of the ras oncogene in the tumorigenesis of Burkitt’s lymphoma has been studied by Seremetis et al. (1989). Introduction of activated N-ras or H-ras oncogenes into EBV immortalized LCLs has led to malignant transformation of these cells. However, their results show that these ras genes are also capable of inducing terminal differentiation of LCLs into plasma cells, and therefore, may have implications in the pathogenesis of terminally differentiated B-lymphoid malignancy such as multiple myeloma rather than in Burkitt’s lymphoma. 30 3.1.2 Other human B-cell Ieukemlas and lymphomas Most of the human B-cell leukemia or lymphoma associated oncogenes that will be described here were identified by molecular cloning of frequent or nonrandom translocation sites, and are recently reviewed by Solomon et al. (1991). It should be noted that unlike the tight association of the c-myc gene with both Burkitt’s lymphoma and murine plasmacytoma, some of the genes described in this section are not invariably found to be activated in all tumors of a particular type. Their impact on the progression of particular neoplasia requires further investigation. B-cell chronic lymphocytic leukemia Four translocations t(1 1 ,14)(q13,q32), t(14,19) (q32,q13), t(8,12)(q24,q22), and t(18;22) have been found in B cell chronic lymphocytic leukemia (B-CLL). The t(11,14)(q13,q32) translocation also occurs in some diffuse small-cell lymphocytic leukemias and diffuse large-cell lymphomas (Yunis, 1983), and multiple myelomas (van der Berghe et al., 1984). The chromosome 11 breakpoints in two CLL patients have been shown to occur only seven nucleotides away from each other, whereas the breakpoint in a diffuse B-cell lymphoma is approximately 0.9 kb distant from those characterized in CLL (T sujimoto et al., 1984a, 1985a). The break point cluster region of this translocation has been denoted as bcI-1. No transcription unit has been detected in this region other than the closely linked PRAD1 gene (Lammie et al., 1991), a gene which was found to be a putative oncogene in parathyroid adenoma (Arnold et al., 1989; Friedman et al., 1990; Rosenberg et al., 1991). PRAD1 encodes a G1 cyclin-like protein (Motokura et al., 1991). Cyclins can form a complex with and activate p34°""2 protein kinase, 31 thereby regulating progress through the cell cycle (for review see Nurse, 1990). PRAD1 mRNA is expressed in many tissues and is highly conserved in bovine and murine tissues (Rosenberg et al., 1991). PRAD1 has been implicated in non- parathyroid neoplasia, squamous cell and mammary carcinomas, and is invariably amplified and overexpressed in these tumors (Lammie et al., 1991). No direct demonstration of the altered expression of PRAD1 has been reported in B-CLL as this thesis is being written. Presumably, the disruption of the cell cycle after alteration of this gene may contribute to the course of B-CLL as in the other tumors. The t(14,19)(q23,q13) translocation involves a deregulation of the col-3 gene (McKeithan et al., 1987; Ohno et al., 1990). As a result of this translocation, chromosome 19 sequences including the bcI-3 gene are juxtaposed to the 5’ end of the 0:1 switch region of the IgH gene on chromosome 14 in a head to head manner. The bcI-3 transcription unit is not disrupted by the translocation, and a more than 3.5 fold increase of the mRNA level was found in total RNA from the peripheral blood of two CLL patients with the t(14;19) translocation as compared to RNA from a patient with the prolymphocytic variant of CLL, which does not contain this translocation. The bcI-3 gene encodes seven tandem copies of the cdc10 motif, a proline rich N-terminal, and a proline-serine rich C-tenninal (Ohno et al., 1990). The cdc10 motif was previously identified in yeast genes that regulate events at the start of the cell cycle (for review see Simanis et al., 1987) and in invertebrate transmembrane proteins involved in cell differentiation pathways (Austin and Kimble, 1987; Seydoux and Greenwald, 1989; Stemberg and Horvitz, 1989). It is not clear which of these two classes of proteins that the bcI-3 gene 32 resembles, but it is clear that it is not a transmembrane protein. The proline rich region has been shown to have transcription activating properties (Mermod et al., 1989). Thus, it is plausible that the bcI-3 gene could be a transcriptional activating factor. The involvement of the bcI-3 gene in B-CLL was later analyzed in a large series of patients by Raghoebier et al. (1991). Unexpectedly, none of the forty four B-CLL studied had a rearrangement within 15 kb of the bcI-3 locus. However, mutations in the bcl-3 gene undetectable in their assays may exist. Whether the bcI-3 gene contributes to the oncogenesis of B-CLL awaits further investigation. The t(8,12)(q24,q22) translocation links c-myc not with an lg enhancer but rather with a locus termed BTG1 on chromosome 12 that presumably deregulates myc (Rimokh et al., 1991). The breakpoint is located in the 3’ end of the myc locus, and increased c-myc expression has been found. Sequences cloned from the breakpoint recognize a 1.8 kb transcript in the CLL cells and in tissues of lymphoid origin. In addition, this chromosome 12 coding sequence is conserved in evolution and a transcript of similar size is present in murine tissues. However, little is known about the function of this putative gene. Whether or not this gene activation represents a general feature for the B-CLL is also unknown. In addition to the possible involvement of the PRAD1, col-3, and c-myc genes in this malignancy, bcI-2 was found rearranged in 3/34 B-CLL through a variant translocation t(18;22). In this translocation, the bcI-2 gene is juxtaposed to the lgtt on: genes in a head to head configuration (Adachi et al., 1990). Bel-2 was also found rearranged in 3/44 B-CLL by Raghoebier and coworkers (1991). The role of bcI-2 in tumorigenesis will be discussed in the following section. The relatively low frequency of any individual oncogene activation in B-CLL U0: Fol WEi 33 may relate to the accuracy of clinical diagnosis, and the broad range of cell types involved in this malignancy. For example, CLL may sometimes be difficult to distinguish from non-Hodgkin’s lymphoma (Bennett et al., 1989; Deegan, 1989). Systemic analyses on a large scale of samples may be required for a better understanding of the molecular basis of this malignancy. Follicular lymphoma The t(14,18)(q32,q21) translocation which occurs in 85% of follicular lymphomas was first described by Fukuhara et al. (1979). The interchromosomal junction has been cloned from several follicular lymphomas (T sujimoto et al, 1985b; Cleary and Sklar, 1985). The recombination region of chromosome 18, bcI-2, is rearranged into the heavy chain enhancer region on chromosome 14 resulting in deregulation of bcI-2 expression (Tsujimoto et al., 1984b, 19850; Cleary and Sklar, 1985; Bakhshi et al., 1985). The normal bcI-2 gene is quiescent is resting B cells, expressed in proliferating B cells, and downregulated in differentiated cells (Graninger et al., 1987; Reed et al., 1987). Inappropriately high levels of bcI-2- immunoglobulin chimeric RNA are present in t(14;18) follicular lymphoma considering their mature B-cell stage (Seto et al., 1988). This indicates that the translocated bcI-2 allele has escaped normal control mechanisms. Nucleotide sequence analysis and biochemical studies from one group suggest that bcI-2 is a GTP-binding protein located on the cytoplasmic surface of cell membranes (Haldar et al., 1989), but a second group has localized it to the inner mitochondrial membrane (Hockenbery et al., 1990). Bel-2 has been shown to prolong cell survival by blocking programmed cell death (Nunez et al., 1990). A similar effect may thus contribute to the formation of follicular lymphomas. Activated bcI-2 genes 34 proved capable of transforming or enhancing the survival of a cultured human B- cell line (T sujimoto, 1989) or NIH3T3 cells (Reed et al., 1988). It is also capable of inducing follicular hyperplasia in transgenic mice (McDonnell et al., 1989) which progresses to a malignant diffuse large-cell lymphoma after a long latency (McDonnell and Korsmeyer, 1991). The long latency, progression from polyclonal to monoclonal disease, and histological conversion, are all suggestive of second alterations. C-myc activation is suggested to be a candidate oncogene both for its occurrence of rearrangement in the diffuse lymphoma, and its well known involvement in B cell neoplasia. In fact, the cooperativity of the c-myc oncogene and 001-2 In tumorigenesis has been shown by Vaux et al. (1988). The authors found the bcI-2 gene can promote the proliferation of bone marrow cells of Eu-myc transgenic mice, some of which become tumorigenic. Another oncogene, c-Ha- ras, was found to complement bcI-2 in malignant transformation of rat embryo fibroblasts (Reed et al., 1990). Pre-B cell acute lymphocytic leukemia Two translocations t(1,19)(q23,p13) and t(5,14)(q31,q32) have been found in pre-B cell acute lymphocytic leukemias (pre-B ALL). The t(1,19)(q23,p13) translocation was described as a common feature for pre-B ALLs (Carroll et al. 1984; Michael et al., 1984; Williams et al., 1984). A fusion protein, E2A-PBX, results from this translocation which links the E2A gene on chromosome 19 to the homeobox containing PBX gene on chromosome 1 (Kamps et al., 1990; Nourse et al., 1990). The E2A gene encodes for two similar immunoglobulin enhancer binding factors, each with a 5’ effector domain and a 3’ DNA binding domain (Murre et al., 1989). The translocation switches the DNA binding domain of the 35 E2A transcription factor with that of PBX, thus placing those genes usually regulated by PBX under the trans-activational control of E2A. Because PBX is not normally transcribed in pre-B cells, the translocation results in ectopic expression of the PBX DNA binding domain and therefore implicates the fusion protein in the tumorigenesis of pre—B cells. The other translocation, t(5,14)(q31,q32) was identified by Grimaldi and Meeker (1989). At the breakpoint of this translocation, interleukin-3 (IL-3) on chromosome 5 is positioned next to the heavy chain enhancer region on chromosome 14 (Meeker et al., 1990). This has led to the hypothesis that the overproduction of IL-3 may result in an autocrine loop that favors leukemogenesis (Meeker et al., 1990). 3.2 Murine plasmacytoma Murine plasmacytoma is induced by the application of a mineral oil such as pristane into the intraperitoneal cavity of a mouse (with a restriction to Balb/c or NZB mice) (Potter and Boyce, 1962; Anderson and Potter, 1969). Balb/c mice develop plasmacytoma with a latency period of 6-12 months after the injection of mineral oil. The long latency has suggested a multi-step nature for the formation of this tumor. Similar to BL, two common characteristics have been associated with almost all plasmacytomas. First, the involvement of a paracrine stimulation is critical for the formation of plasmacytomas. Prior to formation of a plasmacytoma, induction of chronic granuloma tissues that consist primarily of macrophages and neutrophils is found. Granuloma formation has been shown to play an important role both in the 36 development (Potter and MacCardle, 1964) and in the maintenance (Cancro and Potter, 1976) of the primary plasmacytoma. A growth factor (25-kD) that is expressed by a macrophage cell line was found to stimulate the growth of plasmacytoma cells in vitro (Nordan and Potter, 1986). This growth factor is likely to be interleukin-6 (IL-6) as judged by its size and biological activity. lL-6, a pleiotropic cytokine, induces the terminal differentiation of B cells into antibody- secreting cells, and also acts on a variety of other cell types (Kishimoto and Hirano, 1988). Pristane treatment presumably results in the secretion of this growth factor from the chronic granuloma tissue. The involvement of IL-6 in the in vivo growth of plasmacytomas is further implied by the following finding. Overexpression of lL—6 in transgenic mice, although not sufficient to cause the development of mouse plasmacytomas, induces a massive plasmacytosis in thymus, lymph node, spleen, and other tissues (Suematsu et al., 1989). Transfection with the IL-6 gene into lL-6-dependent plasmacytoma cells has been shown to increase their tumorigenicity (Vink et al., 1990). The involvement of growth factor stimulation is similar to that seen in Burkitt’s lymphoma by EBV with the exception of its being paracrine instead of autocrine and its being induced chemically rather than through viral infection. Chromosomal translocation has also been found in almost all plasmacytomas (Yosida et al., 1970; and references listed below). Two types of translocations were first described by Shepard and coworkers (1974a, 1974b, 1978) in transplanted plasmacytoma lines. One consists of a reciprocal translocation between chromosomes 12 and 15, and was later termed O?" vay 0556 THE 195: 37 the "typical" translocation by Ohno et al. (1979) because of its higher occurrence. The other is a reciprocal translocation between chromosomes 6 and 15, and was named the "variant" translocation by the same group because of its relatively lower occurrence. Both translocations are associated with lg gene loci; the "typical“ one usually involves the alpha switch region of the heavy chain gene (Kirsch et al., 1981; Adams et al., 1982; Harris et al., 1982), whereas the "variant“ one involves the kappa light chain gene (van Ness et al., 1983; Webb et al., 1983; Cory et al., 1985). Analyses of the breakpoint on chromosome 15 in these translocations have revealed the involvement of the c-myc gene (Shen-Ong et al., 1982; Adams et al., 1983; Marcu et al., 1983). The consequence of the translocation has been proposed to be similar to that of Burkitt’s lymphoma, i.e., the deregulation of c-myc gene expression (Adams et al., 1983; Bernard et al., 1983). Other secondary events have been observed in some but not all of plasmacytomas. In a few plasmacytomas, activation of the c-mos gene by insertion of an intracisternal A particle element was found in addition to c-myc activation (Rechavi et al., 1982; Canaani et al., 1983; Gattoni-Celli et al., 1983). The v-mos protein exhibit activities of both serine/threonine autophosphorylation (Maxwell and Arlinghaus, 1985) and ATP-dependent DNA binding (Seth et al., 1987) in vitro. Two different biological activities have been associated with the mos protein: inducing monocyte differentiation into macrophages (Kurata et al., 1989) and transforming both NIH3T3 cells and normal kidney cells into malignant cells (Kurata et al., 1987). The biochemical basis of these two different potentialities is not clear. Its cytoplasmic location and kinase activity suggest a role in a signaling pathway. Alteration of a signaling pathway may contribute to the progression of malignancy. Perlmutter and associates (1984) found a t(6,10) recombination besides t(12,15) in the NS-1 murlne plasmacytoma line and another plasmacytoma. Transcription from this locus was detected at a high level, and homologous sequences could be detected in mouse, rabbit, and human. Therefore, it is possible that t(6,10) may encode a gene that plays a role in the course of B cell tumor progression. Lastly, chromosome 11 trisomy is another frequent secondary alteration in murlne plasmacytomas (Ohno et al., 1984). Several oncogenes (Rel, ErbA, ErbB, p53), and cytokine genes (G-CSF, GM-CSF, lL—3, lL-4, IL-5) have been mapped to chromosome 11 (Buchberg et al., 1989; Wilson et al., 1990). Among these genes, lL-4 is a factor involved mainly in the activation of resting B cells, and IL-5 is a factor for the growth and maturation of activated B cells (Kishimoto, 1985). An increased dosage of these two genes may contribute to the formation of plasmacytoma, and perhaps act cooperatively with IL—6. The involvement of multiple oncogenes in the development of murine plasmacytoma is not only suggested by the above findings, but also by the rapid induction of murlne plasmacytoma with viral oncogenes. Infection of mice with Abelson murine leukemia virus after application of pristane greatly reduces the latency of tumor induction (Ohno et al., 1984). These tumors not only express v- abl, but show the same translocations activating c-myc that are observed in tumors induced by pristane alone. Infection with a recombinant retrovirus expressing an avian v-myc after pristane application also accelerated plasmacytoma formation (Potter et al., 1986, 1987). In this case, expression of v-myc seems to replace the 39 requirement for c-myc translocation. As with Burkitt’s lymphoma, no single gene is sufficient for plasmacytoma formation. 3.3 Avian lymphoid leukosis Lymphoid leukosis, a B-cell lymphoma induced by avian leukosis viruses (ALVs), progresses through a series of clinically distinct stages (Cooper et al., 1968). Three stages have been identified in ALV-induced lymphoid leukosis (Cooper et al., 1968; Baba and Humphries, 1985). The earliest detectable lesion is the transformed follicle, a hyperplastic bursal follicle in which the normal follicular architecture is obscured by an abnormal proliferation of lymphoblasts. The lymphoblasts are confined to their follicle of origin during this early stage. Up to 100 transformed follicles may be present in a single bursa, but most of these regress during bursal involution. Then, one or sometimes more than one of the transformed follicles gives rise to a bursal nodule, which is readily identifiable at necropsy. Ultimately, tumor cells from the bursal nodule disseminate to other tissues, resulting in widespread metastasis in the liver, kidney, and spleen. The instance of regression after the first stage suggests the requirement of additional events for further progression. Molecular analyses of ALV-induced lymphomas show that the majority of them contain proviral integrations within or near the c-myc gene resulting in deregulated expression of c-myc (Hayward et al., 1981; Neel et al., 1981; Payne et al., 1982). The insertional activation of c-myc appears to be an early event in lymphomagenesis according to studies of Neiman et al. (1985), but additional proto-oncogene activations are required for progression to the late stages of the 4O disease. Neiman and coworkers have found that HB-1, a v-myc-containing virus, induces transformed follicles when virus-infected cells are used to reconstitute a chemically ablated bursa (Neiman et al., 1985; Thompson et al., 1987). Like ALV- induced transformed follicles, only a small portion of these progress to become lymphomas. These results again indicate that additional genetic events beyond c- myc induction may be required for late stages of tumor progression. Clurman and Hayward (1988) have used a double-infection protocol to facilitate the occurrence of multiple insertional activation in order to identify those genetic events that may function in cooperation with c-myc to induce late stages of progression in ALV- induced lymphomas. In their study, c-myc rearrangement was found in 70% of lymphomas (both primary and metastatic) identified 3 to 4 months after hatching. Additionally, a frequent viral integration locus, c-bic, was found in 14% of the primary lymphomas, and 50% of the metastatic lymphomas. These results confirm that the c-myc gene is frequently a target for insertional activation in ALV-induced lymphomas. They also indicate that c-bic may act synergistically with c-myc during lymphomagenesis. The increased frequency of viral integration at c-bic in metastatic tumors suggests that c-bic is involved in late stages of tumor progression. In summary, studies on oncogene alteration in B-cell lymphomas/leukemias of three species have revealed the mutli-factorial nature of tumor formation. These gene alterations caused by either insertional mutagenesis or chromosomal translocations show different degrees of association with B-cell lymphomas/leukemias. The most commonly seen gene altered, c—myc, shows a more broad range 'of association with tumors of different cell types, whereas other 41 genes altered, such as bcI-2 and IL-6, show a more restricted linkage to lymphoid tumors. The bcI-2 gene alteration has been implicated in lymphoid tumors of multiple developmental stages such as follicular lymphoma, diffuse small and large cell lymphoma, and chronic lymphocytic leukemia. The IL—6 gene alteration, however, couples specifically to plasmacytoma. The significance of these differences is not yet known. Maybe only mature cells have lL-6 receptors. A systemic analysis with various probes on a greater number of tumor samples will be required to obtain a clear sense of the tissue and developmental specificity of these oncogene activations. These affected genes found in B cell neoplasias encode a variety of proteins including growth factors (e.g., IL-6), membrane associated proteins (e.g., N-ras, bcI-2 ), signal transduction proteins (e.g., c—mos), and many cell cycle or differentiation-related regulatory proteins (e.g., c-myc, bcI-I, bcI-3 etc). Since these proteins are presumably involved in the regulation of cell growth and differentiation, deregulation of these genes should be important in the oncogenesis of B-cell lymphomas/leukemias. Although their roles in these malignant tumors seem obvious, a direct test of their oncogenic potential should be provided to ultimately distinguish their causative effects in tumorigenesis from their simply being the result of malignancy. Furthermore, analyses of end point specimens, such as malignant tumors, do not provide direct information on the effects of differentiation upon oncogene activation or the nature of genes that may be activated in lymphomas/leukemias in addition to those in an obvious translocation. To address these questions, a few model systems, including the use of in vitro culture systems, have been used. Results of these studies are summarized in the 3.4 I rm: Us vim TECE “Mp V-at 42 following section. 3.4 In vitro transformation of murlne B cells A number of oncogenes or growth factors have been found to induce transformation of B lymphoid cells in vitro. These may include (i) oncogenes of the ras oncogene family which code for protein with GDP-binding activity (for review see Barbacid, 1987), (ii) oncogenes of the src oncogene family which code for tyrosine-specific protein kinases (for review see Hunter and Cooper,1985), (iii) the v-fms oncogene whose cellular counterpart encodes a colony stimulating factor-1 receptor, and (iv) the interleukin-7 (IL-7) gene which codes for a pre-B cell growth factor gene (Namen et al., 1988). V-abI is the first viral oncogene that is capable of transforming murlne pre-B cells in vitro (Sklar et al., 1974; Rosenberg et al., 1975; Rosenberg and Baltimore, 1976). Infection of cell cultures derived from murlne fetal liver, bone marrow or spleen, but not thymus with Abelson murine leukemia virus (A-MuLV) carrying the v-abl oncogene give rise to permanently growing, neoplastic cell lines. These in vitro isolated cell lines are similar morphologically to A-MuLV-induced tumor cells display a characteristic of B cells, and have been found to be preferentially pre-B cells (Siden et al., 1979). Another oncogene of the src gene family, v-fes, also exhibits oncogenic potential on murine pre-B cells in vitro (Pierce and Aaronson, 1983). In this study, bone marrow cells were infected with a v-fes containing retrovirus (Snyder-Theilen feline sarcoma virus, ST-FeLV) and selected for their ability to grow on soft agar, a characteristic of transformed cells. In both cases, A-MuLV or ST-FeLV infection alone appears to be sufficient to induce aggressive If I‘ll". re. ‘U S" “U U tie 43 tumors in syngeneic mice with a 1-3 weeks latency. However, results from Whitlock and Witte (1981) using a fresh bone marrow cell suspension as target for the A-MuLV infection and a feeder layer to support the growth of the initial infected population, show that the A-MuLV infected cells are initially poorly oncogenic and that they become progressively tumorigenic and growth independent only after progression on normal adherent bone marrow feeder layers. This latter result suggests that a minor subpopulatlon capable of unrestricted growth is present at the initiation of the culture and gradually becomes the predominant population through a slight growth advantage. Alternatively, the A-MuLV-infected cells may undergo secondary changes which further alter their growth properties. Using clonal cell lines isolated from the A-MuLV infected cells, Whitlock et al. (1983b) have shown that the progression of these clonal cell lines to a more malignant growth phenotype occurs with no changes in viral related properties. The expression of cellular genes appears to alter the growth properties of lymphoid cells after their initial transformation by A-MuLV. The discrepancy between Rosenberg’s and Whitlock’s results may residue in the properties of cells used for the initial infection, and the stringency of the growth parameters used to determine transformation. Since the cells used by Rosenberg’s group have been cultured in vitro for a period of time, cells with a slight growth advantage may be preselected for viral infection. The lack of a feeder layer again selects for cells with a more transformed phenotype. A similar explanation may be applied to the results from Pierce and Aaronson (1983), in which the soft agar assay may select for events additional to the ST-FeLV infection. In conclusion, these results suggest that v-abl or v-fes may, by itself, be sufficient to initiate transformation of B cells but may 44 require additional events, such as the activation of cellular oncogenes, for expression of the fully transformed state. The involvement of the v-Ha-ras and v-bas oncogenes in the in vitro transformation of lymphoid cells was demonstrated by Pierce and Aaronson (1982; 1984) using a similar approach as that described for the v-fes oncogene (see above). The Harvey murine sarcoma virus carrying a v-Ha-ras oncogene, and the Balb-murine sarcoma virus carrying a v-bas oncogene were used instead. The resulting transformed cell lines were as tumorigenic as those infected by A-MuLV. Cells transformed by v-Ha-ras or v-bas displayed characteristics of immature lymphoid cells: high terminal deoxynucleotidyl transferase (T dt) activity, the presence of F0 receptor, and mercaptoethanol dependence for growth. The lack of u chain expression, and of Thy-1 antigen on these transformed cells suggested that these cells might be at an earlier stage of differentiation than pre-B or pre-T lymphoid cells. Holmes et al. (1986) further confirmed the role of oncogenes in the ras gene family in the in vitro transformation of B cells. They demonstrated that transformed cell lines obtained from cells infected with viruses containing v-Ha-ras, v-bas, and v-K-ras display characteristics of pro-B and pre-B cells. Their finding suggest that a wide range of oncogene can induce B cell transformation in vitro. The difference in the target cells for the ras oncogene family in the results of Holmes et al. and, Pierce and Aaronson may be a reflection of a continuous differentiation, which may occur under the influence of the microenvironment of a particular experimental setup, of a lymphoid progenitor (or a hematopoietic progenitor in a broader aspect). This hypothesis is supported by the isolation of Will a i1 01 DIE A3; by *0 O. 45 macrophage-like cell lines from pre-B cell lines containing the v-Ha-ras oncogene obtained in the same study (Holmes et al., 1986). This lineage-switch phenomenon is also found in pre-B cells transformed by the v-fms oncogene (Borzillo et al., 1990; see below), and the v-raf oncogene (Klinken et al., 1988), as well as in our study (Appendix A). Another disparity is that viruses carrying v-Ha-ras, v—bas, and v-Ki-ras can also induce sarcomas and erythroleukemias in susceptible animals (Peters et al., 1974; Harvey, 1964; Scher et al., 1975; Hankins and Scolnick, 1981) and transform myeloid cells in vitro (Pierce and Aaronson, 1985), whereas A-MuLV seems to have a restricted target cell for transformation. This tissue-preference of tumorigenesis of A-MuLV appears to be directly associated with the v-abl oncogene from the studies of transgenic mice in which tumors of a B lymphoid origin are found predominately in transgenic mice carrying the v-abl oncogene (for review, see Adams and Cory, 1991). These results suggest that the tyrosine kinase encoded by the v-abl oncogene may belong to a pre-B cell specific signal transduction pathway, whereas the G-protein activity encoded by the ras gene family may represent a common signal pathway shared by several cell types. Borzillo and Sherr (1989) have shown that the v-fms gene is capable of inducing transformation of pre-B cells. As with v-abl transformants, only the late- passage cultures give rise to factor-independent variants that proliferate in the absence of feeder layers, and become tumorigenic in syngeneic mice. The v-fms oncogene encodes an analog of CSF-1R that retains a functional ligand-binding domain but acts constitutively as a tyrosine kinase because of mutations. The mechanism of this mononuclear-macrophage—lineage related receptor functioning 46 in pre-B cells is not well known. The fact that some of the v-fms transformed pre-B cell line may undergo a lineage switch to macrophages when transferred from RPMI1640 to Iscove modified Dulbecco medium suggests that v-fms or its cellular analogue may be a common intermediate in the signal transduction pathways of both the lymphoid and myeloid differentiation (Borzillo et al., 1990). In addition to the above oncogene, a pre-B cell growth factor, IL-7 also shows transforming potential on pre-B cells (Young et al., 1991; Overell et al., 1991). However, the low cloning frequency of IL-7 transformants and the long latency for tumorigenesis suggest that additional events are required in generating a fully transformed phenotype. In summary, a variety of oncogenes, growth factors or receptors have been implicated in the in vitro transformation of B cells as in the in vivo tumorigenesis of B cell tumors from different species. Consistent with the multi-step tumorigenesis dogma, none of them are tumorigenic by themselves, and additional events must exist. These results validate the use of the in vitro model to examine the events that underlie tumor progression. In my thesis, oncogene cooperation was used as a model to define the multi-factorial molecular events in the tumor progression of murine pre-B cells. The advantage of using oncogene cooperativity as a model to dissect the events contributing to tumor progression is the enhanced likelihood of identifying genes that by themselves are not capable of demonstrable transformation in growth factor independence assays, soft agar assays, and tumor challenges. However, genes with a subtle or negligible phenotype in the above assays may be able to transform 8 cells in cooperation with other oncogenes in a synergistic manner, for example 47 with v-myc or v-Ha—ras, and thus are potentially involved in the course of tumor progression. In setting up a system to examine cooperative transformation of B lymphoid cells, v-myc was the first candidate because c-myc activation was found in many of the B cell malignancies described above. Unfortunately, the v-myc gene was not able by itself to transforme primary bone marrow cells in the Whitlock and Witte culture system and may be lethal (data not shown, Stevenson and Volsky, 1986). We therefore chose v-Ha-ras, partly because v-Ha-ras has been found to transform murlne B cells into an intermediate transformed phenotype (Schwartz et al., 1986a; Holmes et al., 1986), and partly because ras gene aberration was found in some 8 cell neoplasias (see previous section). The two model systems used in my studies are described in the “Introduction". 48 Literature cited: Adachi M., A. Tefferi, P.R. Greipp, T.J. Kipps, and Y. Tsujimoto. (1990). Preferential linkage of bcI-2 to immunoglobulin light chain gene in chronic lymphocytic leukemia. J. Exp. Med. 171, 559-564. Adams J.M., and 8. Cory. (1991). Transgenic models of tumor development. Science 254, 1161-1167. Adams J.M., E. Webb. J. Mitchell, 0. Bernard, and S. Cory. (1982). Transcriptionally active DNA region that rearranges frequently in murlne lymphoid tumors. Proc. Natl. Acad. Sci. USA 79. 6966-6970. Adams J.M., S. Gerondakls, E. Webb, LM. Corcoran, and S. Cory. (1983). Cellular myc gene is altered by chromosome translocation to an immunoglobulin locus in murlne plasmacytomas and Is rearranged similarly in human Burkitt’s lymphomas. Proc. Natl. Acad. Sci. USA 80. 1982-1986. Adams J.M., A.W. Harris, C.A. Pinkert, LM. Corcoran, W.S. Alexander, S. Cory, R.D. Palmiter, and BL Brlnster. (1985). The c-myc oncogene driven by lmmunoglobulln enhancers Induce lymphoid malignancy in transgenic mice. Nature 318, 533-538. Arnold A., H.G. Kim, R.D. Gaz, P.L. Eddy, Y. Fukushima, MG. Byer, TB. Shows, and HM. Kronenberg. (1989). Molecular cloning and chromosomal mapping of DNA rearranged with the parathyroid hormone gene in a parathyroid adenoma. J. Clin. Invest. 83, 2034-2040. Austin J.. and J. Kimble. (1987). ng-t is required in the germ line for regulation of the decision between mitosis and meiosis in C. elegans. Cell 51, 589-599. Barbacld M. (1987). Fias genes. Ann. Rev. Biochem. 56, 779-827. Bargmann C.I., M.-C. Hung, and RA Weinberg. (1986). The neu oncogene encoded an epldennal growth factor receptor-related protein. Nature 319, 226-230. Bates G.P., B.J. Wainwright, R. Williamson, and S.D.M. Brown. (1986). Microdissection and microcloning from the short arm of human chromosome 2. Mol. Cell. Biol. 6, 3826-3830. Baumbach W.R., EM. Colston, and MD. Cole. (1988). Integration of the Balb/c ecotropic provlms Into the colony-stimulating factor-1 growth factor locus in a myc retrovirus induced murlne monocyte tumor. J. Virol. 62, 3151-3155. Bennett J.M., D. Catovsky, M.T. Daniel, G. Flandrin. D.A.G. Galton, H.R. Gralnick and C. Sulatn. (1989). Proposal for the classification of chronic (mature) B and T lymphoid leukemias and lymphomas. J. Clin. Pathol. 42, 567. Bernard 0., S. Cory, S. Gerondakls, E. Webb, and J. M. Adams. (1983). Sequence of the murlne and human cellular myc oncogene and two modes of myc transcription resulting from chromosome translocation in B lymphoid tumours. EMBO J. 2, 2375-2383. Birchmeier C., D. Broek, T. Toda, S. Powers, T. Kataoka, and M. Wigler. (1985). Conservation and divergence of RAS protein function during evolution. Cold Spring Harb. Symp. Quant. Biol. 50, 721 -725. Bonner T.l., C. O’Connell, and M. Cohen. (1982). Cloned endogenous retrovlral sequences from human DNA. Proc. Natl. Acad. USA 79. 4709-4713. 49 Borzlllo G.V., and OJ. Sherr. (1989). Early pre-B-cell transformation Induced by the v-fms oncogene In long-term mouse bone marrow cultures. Mol. Cell. Biol. 9, 3973-3981. Borzillo G.V., R.A. Ashman, and OJ. Sherr. (1990). Macrophage lineage switching of murlne early pre-B lymphoid cells expressing transduced fms genes. Mol. Cell. Biol. 10, 2703-2714. Breuer M.C., H.T. Cuypers, and A. Bems. (1989a). Evidence for the Involvement of pIm-2 a new common proviral insertion site in progression of lymphomas. EMBO J. 8, 743-747. Buchberg A.M., E. Brownell, S. Nagata, N.A. Jenkins, and MG. Copeland. (1989). A comprehensive genetic map of murlne chromosome 11 reveals extensive lineage conservation between mouse and human. Genetics 122, 153-161 . Burkitt DP. (1958). A sarcoma involving the jaws in African children. Br. J. Surg. 46, 218-223. Canaani E., O. Dreazen, A. Klar, Rechave, D. Ram, J.B. Cohen, and 0. leol. (1983). Activation of the c-mos oncogene in a mouse plasmacytoma by insertion of an endogenous intracisternal A-particle genome. Proc. Natl. Acad. Sci. USA 80, 7118-7122. Cancro M. , and M. Potter. (1976). The requirement of an adherent substratum for the growth of developing plasmacytoma cells in vivo. J. Exp. Med. 144, 1554-1566. Carroll A.J., W.M. Crist, R.T. Parmley, M. Roper, M.D. Cooper, and WM. Finley. (1984). Pre-B cell leukemia associated with chromosome translocation 1:19. Blood 63, 721-724. Chaudhuri J.P., W. Vogel, I. Volculescu, and U. Wolf. (1971). A simplified method of demonstrating Glemsa band pattern in human chromosomes. Human Genetlk 14, 83. Chen S.J., N.J. Holbrook, KF. Mitchell, C.A. Vallone, J.S. Greengard, G.R. Crabtree, and Y. L]. (1985). A viral long terminal repeat In the Interleukin-2 gene of a cell line that constitutively produces interleukin-2. Proc. Natl. Acad. Sci. USA 82, 7284-7288. Clurman BE. and W.S. Hayward. (1989). Multiple proto-oncogene activation in avian leukosis virus-induced lymphomas: evidence for stage-specific events. Mol. Cell. Biol. 9, 2657-2664. Cole J.M., C. Righi. C. Dissous, G. Gegonne, and D. Stehelin. (1983). Molecular cloning of the avian acute transforming retrovirus MH2 reveals a novel cell-derived sequence(v-mll) In addition to the myc oncogene. EMBO J. 2, 2189-2194. Collins 8., and M. Groudine. (1982). Amplificatlon of endogenous myc-related DNA sequences in a human myeloid leukemia cell line. Nature 298, 679-681. Cooper M.D., LE. Payne, P.B. Dent, B.P. Burmester, and RA. Good. (1968). Pathogenesis of avian leukosis. J. Natl. Cancer Inst. 41, 374-389. Cooper 08., M. Park, D.G. Blair, M.A. Tainsky, K. Huebner, CM. Croce, and G.F. Vonde Woude. (1984a). Molecular cloning of a new transforrnlng gene from a chemically transformed human cell line. Nature 311, 29-33. Croce CM, and RC. Nowell. (1986). Molecular genetics of human B cell neoplasia Adv. Immunol. 38, 245. Croce C.M., M. Shander, J. Martinis, L Clcurel, G.G. D’Ancona T.W. Dolby. and H. Koprowskl. (1979). chromosomal location of the genes for human immunoglobulin heavy chains. Proc. Natl. Acad. Sci. USA 76, 3416-3419. 50 Cuypers H.T., G. Selten, W. Quint, M. Zijlstra, ER. Maandag, W. Boelens, P. van Wezenbeek, C. Melief, and A Bems. (1984). Murine leukemia virus-induced T cell lymphomagenesis: integration of proviruses in a distinct chromosomal region. Cell 37, 141-150. Delll Bovi P., A.M. Curatola, F.G. Kern, A. Greco, M. Ittmann, and C. Basilica. (1987). An oncogene ls isolated by transfection of Kaposi’s sarcoma DNA encoded a growth factor that is a member of the FGF family. Cell 50, 729-737. Der C.J., T.G. Krontlris, and GM. Copper. (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. 3637-3640. de The’ G., A Geser, N.E. Day. P.M. Tukel, E.H. Williams, D. Beri, P.G. Smith, AG. Dean, G,W. Bornkamm, P. Feorlno, and W. Henle. (1978 ). Epidemiological evidence for a causal relationship between Epstein-Barr Virus and Burkitt's lymphoma: result of Uganda prospective study. Nature 274, 756-761. Diamond L. ,T.G. O’Brien, and WM. Baird. (1980). Tumor promoters and the mechanism of tumor promotion. Adv. Can. Res. 32, 1-74. Dickson C., R. Smith, S. Brookers. and G. Peters. (1984). Tumorigenesis by mouse mammary tumor virus: proviral activation of a cellular gene in the common integration region int-2. Cell 37, 529-536. DiPaolo J.A. (1983). Relative difficulties In transforming human and animal cells In vino. J. Natl. Cancer Inst. 70, 3-7. Duyk G.M., S. Kim, R.D. Myers, and DR. Cox. (1990). Exon trapping: a genetic screen to Identify candidate transcribed sequences in cloned mammalian genomic DNA Proc. Natl. Acad. Sci. USA 87, 8995-8999. Edstrom J.-E., R. Kaiser, and D. Rohme. (1986). Microcloning of mammalian metaphase chromosomes. Methods Enzymol. 151, 503-516. Erikson J., J. Martinis, and CM. Croce. (1981). Assignment of the genes for human lambda lmmunoglobulln chains to chromosome 22. Nature 294, 173-175. Eva. A.,, and SA. Aaronson. (1985). Isolation of a new human oncogene from a diffuse B-cell lymphoma. Nature 316, 273-275. Fasano D. , D. Blmbaum, L Edlund, J. Fogh, and M. Wigler. (1984). New human transforrnlng genes detected by a tumorigenecity assay. Mol. Cell. Biol. 4, 1695-1705. Fearon ER, and B. Vogelstein. (1990). A genetic model for colorectal tumorigenesis. Cell 61, 759-769. Fisher J.C., and J.H. Holloman. (1951 ). A hypothesis for the origin of cancer loci. Cancer 4, 916-918. Foulds L (1975). Neoplastic development. Vol. II. (New York: Academic press). Fung Y.-KT., W.L Louis, LB. Crittenden. and H.J. Kung. (1983). Activation of the cellular oncogene c-erbB by LTR Insertion: Molecular basis of erythroblastosls by Avian Leukosis Virus. Cell 33, 357-368. Gabrielson E.W., and CC. harrls. (1985). Use of cultured human tissues and cells In carcinogenesis research. J. Cancer Res. Clin. Oncol. 110, 1-10. 51 Gaidano G.. P. Ballerini, J.Z. Gong, G. lnghlraml, A Neri, E.W. Newcomb, I.T. Magrath, D.M. Knowles, and R. Dalia-Favera. (1991). p53 mutations In human lymphoid malignancies: Association with Burkitt lymphoma and chronic lymphocytic leukemia. Proc. Natl. Acad. Sci. USA 88, 5413-5417. Garcia M., R. Wellinger, A. Vessaz, and H. Diggelmann. (1986). A new site of Integration for mouse mammary tumor virus proviral DNA common to Balb/cf(CSH) mammary and kidney adenocarcinomas. EMBO J. 5, 127-134. Geroge D.L, B. Glick, S. Trusko, and N. Freeman. (1986). Enhanced c-Kl-ras expression associated with Friend Virus Integration is a bone-marrow derived mouse line. Proc. Natl. Acad. Sci. USA 83, 1651-1655. Geser A., G. Lenoir, M. Anvret, G.W. Bornkamm, G. Klein, E.H. willlams, D.H. Wright, and G. de The' (1983). Epstein-Barr Virus markers in a series of Burkitt's lymphoma from West Nile District of Uganda. European J. Cancer. Clin. Oncol. 19, 1394-1404. Gordon J., S.C. Ley, M.D. Melamid, LS. English, and NC. Hughes Jones. (1984). lmmortallzed B lymphocytes produces B cell growth factor. Nature 310, 145-147. Graham FL, and A.J. van der Eb. (1973). Transformation of rat cells by DNA of human adenovirus 5. Virology 54, 536-539. Graninger W., M. Seto, B. Boutain, P. Goldman, and SJ. Korsmeyer. (1987). Expression of bcI-2 and bcI-2-Ig fusion transcripts in normal neoplastic cells. J. Clin. invest. 80, 1512-1515. Grazianl D, D. Ron, A. Eva, and K. Srivastavas. (1989). The human dbl-protooncogene product is a cytoplasmic phosphoprotein which Is associated with the cytoskeletal matrix Oncogene 4, 823-829. Grimaldi J.C., and TC. Meeker. (1989). The t(5;14) chromosomal translocation In a case of acute lymphocytic leukemia joins the interleukin-3 gene to the immunoglobulin heavy chain gene. Blood 73, 2081 -2085. Haldar S., C. Beatty, Y. Tsujimoto. and CM. Croce. (1989). The bcI-2 gene encodes a novel G protein. Nature 342, 195-198. Halevy 0., D. Michalovitz, and M. Oren. (1990). Different tumor-derived p53 mutants exhle distinct biological activities. Science 250, 113-116. Harris H. (1988). The analysis of malignancy by cell fusion: the position in 1988. Cancer Res. 48, 3302-3306. Hayward W., 8.6. Neel, and S. Astrin. (1981). Activation of a cellular one gene by promoter Insertion in ALV-Induced lymphoid leukosis. Nature 290, 475-480. Hicks G.G.. and M. Mowat. (1988). Integration of Friend Murine Leukemia Virus Into both alleles of the p53 oncogene In an erythroleukemia cell line. J. Virol. 62, 4762-4755. Hockenbery D., G. Nunez, C. Milliman, R.D. Schreiber, and J. Korsmeyer. (1990). Bcl-2 Is an Inner mitochondrial membrane protein that blocks programmed cell death. Nature 348, 334-336. Holmes KL, J.H. Pierce, W.F. Davison, and HG Morse, III. (1986). Murine hematopoietic cells with pre-B or pre-B/myelold characteristics are generated by In vitro transformation with retrovirus containing fes, ras, abI, and src oncogenes. J. Exp. Med. 160, 443-459. Hunter T. (1991). Cooperation between oncogenes. Cell 64, 249-270. swim“ New. lame Id be m 52 Hunter T., and J.A. Cooper. (1985). Protein tyrosine kinases. Ann. Rev. Biochem. Iman, OS. and CC. Harris. (1991). Crit. Rev. Oncogen. 2, 161. lhle J.N., 8.8. White, B. Sisson, D. Parker, D.G. Blair, A. Schultz, C. kozak, RD. Cunsford. D. Askew, Y. Weinstein, and R.J. lsofort. (1989). Activation of c-Ha-ras protooncogenes by retrovirus Insertion and chromosomal rearrangement In a Moloney leukemia virus-Induced T cell leukemia. J. Virol. 63, 2959-2966. Kamps M.P., C. Murre, X.-H. Sun, and D. Baltimore. (1990). A new homeobox gene contributes the DNA binding domain of the t(1;19) translocation protein in pre-B ALL Cell 60, 547-555. Kan W.C., C.S. Flordellis, C.F. Garon, P. Duesberg, and TS. Papas. (1983). Avian carcinoma virus MH2 contains a transformation-specific sequence, mht, and share the myc sequence with M029, CMII, and OKIO viruses. Proc. Natl. Acad. Sci. USA 80, 6656-6670. Kirsch I.R., J.V. Ravetch, S.-P. Kwan, E.E. Max, R.L Ney, and P. Leder. (1981). Mdtlple lmmunoglobulln switch region homologies outside the heavy chain constant region locus. Nature 293. 585-587. Klshimoto T. (1985). Factors affecting B-ceII growth and differentiation. Ann. Rev. Immunol. 3, 133-157. Klshimoto T., and T. Hirano. (1988). Molecular regulation of B Iyrnphocyte Response. Ann. Rev. Immunol. 6, 485-512. Kitayama H., Y. Sugimoto, T. Matsuzakl, Y. Ikawa. and M. Noda. (1989). A ras related gene with transformation suppressor activity. Cell 56, 77-84. Klein 6.. and E. Klein. (1985). Evolution of tumors and the impact of molecular biology. Nature 315, 190-195. Klein E., G. Klein, J.S. Nadkarni, J.J. Nadkaml, H. ngzell, and P. Clifford. (1967). Surface lgM specificity on cells derived from a Burkitt’s lymphoma. Lancet 2, 1068-1070. Klinken S.P., W.S. Alexander, J.M. Adams. (1988). Hemopoletlc lineage switch: v-raf oncogene converts Eu-myc transgenic B cells into macrophages. Cell 53, 857-867. Knudsen AG., Jr., and AT. Meadows . (1980). Regression of neuroblastoma IV-S: a genetic hypothesis. N. Engl. J. Med. 302, 1254-1256. Kurata S., N. Kurata, and Y. Ikawa. (1987). Production of recombinant rat vlmses as a method of oncogene Isolation in coculture medium. Cancer Res. 47, 5908-5912. Kurata N., H. Aklyama, T. Tanlyama, and T. Marunouchi. (1989). Dose-dependent regulation of macrophage differentiation by mos mRNA In a human monocytlc cell line. EMBO J. 8, 457-463. Lamb P., and L Crawford. (1986). Characterization of the human p53 gene. Mol. Cell. Biol. 6, 1379-1385. Lammie G.A., V. Fantl, R. Smith, E. Schuuring, S. Brookes, R. Mlchalides, C. Dickson, A. Arnold, and G. Peters. (1991 ). D1 13287, a putative oncogene on chromosome 11q13, is amplified and expressed in squamous cell and mammary carcinomas and linked to BCL-I. Oncogene 6, 439-444. 53 Lenoir G.M., T. Philip, and R. Sohier. (1984a). Burkitt-type lymphomas-EBV association and cytogenetic markers In cases from various geographic locations. In Pathogenesis of Leukemlas and lymphomas: environmental influences. I.J. Magrath, G.T. O’Conor, and B. Ramot. eds. (New York: Raven Press). pp. 283-295. Little C.D., D.N. Carney, and AF. Gazdar. (1983). Amplificatlon and expression of the c-myc oncogene In human lung cancer cell lines. Nature 306, 194-196. Lombardi L, E.W. Newcomb, and R. Dalia-Favera. (1987). Pathogenesis of Burkitt lymphoma: expression of an activated c-myc oncogene causes the tumorigenic conversion of EBV-Infected human B lymphoblasts. Cell 49, 161-170. Lfidecke H.-J., G. Senger, U. Claussen, and B. Horsthemke. (1989). Cloning defined regions of the human genome by microdissection of banded chromosomes and enzymatic amplification. Nature 338. 348-350. Malcolm 8., P. Barton, C. Murphy, M.A. Ferguson-Smith, D.L Bentley, and TH. Rabbltts . (1982). Localization of human immunoglobulin K chain variable region to the short arm of chromosome 2 by in situ hybridization. Proc. Natl. Acad. Sci. USA 79, 4957-4961. Marth J.D., R. Reet, E.G. Krebs, and RM. Pertmutter. (1985). A lymphocyte-specific protein tyrosine kinase gene is rearranged and overexpressed in murine T cell lymphoma LSTRA. Cell 43, 393-404. Martin-Zanaca D., S.H. Hughes, and M. Barbacid. (1986). A human oncogene formed by the fusion of truncated tropomyosin and protein tyrosine kinase sequences. Nature 319, 743-748. Maxwell SA, and RB. Arlinghaus. (1985). Serine kinase activity associated with Moloney murlne sarcoma virus-124-encoded p37 mos. Virology 143, 321-333. McDonnell T.J., and SJ. Stanley. (1991). Progression from lymphoid hyperplasia to high-grade malignant lymphoma in mice transgenic for the t(14;18). Nature 349, 254-256. McDonnell T.J., N. Deane, F.M. Platt, G. Nunez, U. Jaeger, J.P. Mckeam, and SJ. Korsmeyer. (1989). bcI-2-Immunoglobulin transgenic mice demonstrate extended 8 cell survival and follicular Iymphoproliferation. Cell 57, 79-88. McKeithan T.W., J.D. Rowley, T.B. Shows, and MO. Diaz. (1987). Cloning of the chromosome translocation breakpoint junction of the t(14;19) In chronic lymphocytic leukemia. Proc. Natl. Acad. Sci. USA 84. 9257-9260. Meeker T.C., D. Hardy, C. Willman, T. Hogan, and J. Abrams. (1990). Activation of the Interleukin-3 gene by chromosome translocation in acute lymphocytic leukemia with eosinophllia Blood 76, 285-289. Menezes J., W. Leibold, and G. Klein. (1975). Biological differences between Epstein-Barr Virus strains with regard to lymphocyte transforming ability, superlnfection and antigen induction. Exp. Cell. Res. 92. 478-484. Mermod N., E.A. O’Nell. T.J. Kelly, and R. Tijan. (1989). The proline-rich transcriptional activator of CTF/NF-I is distinct from the replication and DNA binding domain. Cell 58, 741 -753. Mltelman F. (1991). Catalogue of chromosome aberrations In cancer. In Progress and Topics In Cytogenetlcs. Vol. 5, AA. Sandberg. ed. (New York: WIIey-Liss Press). Moreau-Gacheiin F., A Tavitian, and P. Tambourln. (1988). Spi-I Is a putative oncogene in virally induced murlne erythroleukemias. Nature 331, 277-280. 54 Morishita K, 0.8. Parker, M. Mucenski, NA Jenkins, N.G. Copeland; and J.N. lhle. (1988). Retroviral activation of a novel gene encoding a zinc finger protein in lL-3 dependent leukemia cell lines. Cell 59, 831-840. Motokura T., T. Bloom, H.G. Kim, H. Juppner, J.V. Ruderrnan, HM. Kronenberg. and A Arnold. (1991). A novel cyclin encoded by a bcI-1 linked candidate oncogene. Nature 350, 512-515. Mucenski M.L., B.A. Taylor, J.M. lhle. J.N. Hartley, H.C. Morse, lli, N.A. Jenkins, and MG. Copeland. (1988a). identification of a common ecotropic viral integration site evi-i in the DNA of AKXD murlne myeloid tumors. Moi. Cell. Biol. 8, 301-308. Murray M.J.. J.M. Cunningham, LF. Parada, F. Daultry, P. Lebowitz, and RA Weinberg. (1983). The HL-60 transforming cell sequence: a ras oncogene coexisting with altered myc gene in hematopoietic tumor. Cell 33, 749-757. Murre C., P.S. McCaw, H. Vaessin, M. Caudy, LY. Jan, Y.N. Jan, C.V. Cabrera, J.N. Buskln, S.D. Hauschka, AB. Lassar, H. Weintraub, and D. Baltimore. (1989). Interactions between heterologous helix-ioop-helbi proteins generates complexes that bind specifically to a common DNA sequence. Cell 58. 537-544. Namen A.E., A.E. Schmierer, C.J. March, R.W. Overell. LS. Park, D.L Urdal, and D.Y. Mochizukl. (1988). B cell precursor growth-promoting activity: purification and characterization of a growth factor active on lymphocyte precursors. J. Exp. Med. 107, 988-1002. Neil J.C., R. McFarlane, N.M. Wilkie, D.e. Onions, G. Lees, and O. Jarett. (19884). Transduction and rearrangement of the myc gene by feline leukemia virus in naturally occurring T -cell leukemias. Nature 308, 814-820. Neiman P.E., C. Wolf, P.J. Enrletto, and GM. Cooper. (1985). A retrovlral myc gene induces preneoplastic transformation of lymphocytes in a bursal transplantation assay. Proc. Natl. Acad. Sci. USA 82, 222-226. Nilsson K, and G. Klein. (1982). Phenotyplc and cytogenetic characteristics of human B-lymphold cell lines and their relevance for the etiology of Burkitt’s lymphoma. Adv. Cancer. Res. 37, 31943). Noguchi T., F. Galland, M. Batoz, M.-G. Mattel, and D. Bimbaum. (1988). Activation of a mcf.2 oncogene by deletion of amino-terminal coding sequences. Oncogene 3, 709-715. Nordan RP, and M. Potter. (1986). A macrophage-derived factor required by plasmacytomas for survival and proliferation In vitro. Science 233, 566-569. Nowell PC, and DA Hungeriord. (1960). A minute chromosome in human chronic grandocytic leukemia . Science 132, 1497. Nunez G., L London, D. Hockenbery, M. Alexander, J.P. McKeam, and SJ. Korsmeyer. (1990). Deregulated bcI-2 gene expression selectively prolongs survival of growth factor-deprived hematopoietic cell lines. J. lmmunoi. 144, 3002-3010. Nunn M.F., P.H. Seeberg, C. Moscovici, and P. Duesberg. (1983). Tripartite structure of the avian erythroblastosls virus E26 transforming gene. Nature 306, 391-393. Nusse R., and HE. Varmus. (1982). Many tumors induced by the mouse mammary tumor virus contain a provirus integrated in the same region of the host genome. Cell 31, 99-109. Nusse R., A. van Ooyen, D. Cox, Y.K.T. Fung, and H. Varmus. (1984). Mode of proviral integration of a putative mammary oncogene(int-1) on chromosome 15. Nature 307, 131-136. 55 Ochiya T., A. Fujiyama, S. Fukushige, i. Hatakada, K Matsubara. (1986). Molecular cloning of an oncogene from a human hepatocellular carcinoma. Proc. Natl. Acad. Sci. USA 83, 4993-4997. O'Conor G.T., H. Rappaport, and EB. Smith. (1985). Childhood lymphoma resembling Burkitt tumor in the United States. Cancer 18, 411-417. Ohno S., M. Babonits, F. Wiener, J. Spira, G.Klein, and M. Potter. (1979). Nonrandom chromosome changes including the lg gene-carrying chromosomes 12 and 6 in pristane-induced mouse plasmacytomas. Cell 18, 1001-1007. Ohno S., S. Migita, F. Wiener, M. Babonits, G. Klein, J.F. Mushinski, and M. Potter. (1984). Chromosomal translocations activating myc sequences and transduction of v-abl are critical events in the rapid induction of plasmacytomas by pristane and Abelson Virus. J. Exp. Med. 159, 1762-1777. Ohno H., G. Takimoto, and T.W. McKeithan. (1990). The candidate proto-oncogene bcI-3 is related to genes implicated in cell lineage determination and cell cycle control. Cell 60, 991-997. Padua 6.8., N. Barrass, and GA. Currie. (1984). A novel transforming gene in a human malignant melanoma cell line. Nature 311, 671-673. Palmiter RD, and R.L. Brinster. (1986). Germ-line transformation of mice. Ann. Rev. Genet. 20, 465-499. Payne G.S., J.M. Adams, and HE. Varmus. (1982). Multiple arrangements of viral DNA and an activated host oncogene in bursal lymphomas. Nature 295, 209-213. Perlmutter R.M., J.L Kiitz, D. Pravtcheva, F. Ruddle. and L Hood. (1984). A novel 6:10 chromosomal translocation in the murlne plasmacytoma NS-1. Nature 307, 473-476. Peters R.L, LS. Rabstein, R. van Vleck, G.J. Keiloff, and R.J. Huebner. (1974). Naturally occurring sarcoma viruses of the Baib/cCr mouse. J. Natl. Cancer inst. 53, 1725. Peto R., F.J. Roe, PM Lee, L. Levy, and J. Clark. (1975). Cancer and ageing in mice and men. Br. J. Cancer 32, 411-426. Pierce J.H., and S.A. Aaronson. (1983). In vitro transformation of murlne pre-B lymphoid cells by Snyder-Theiien feline sarcoma virus. J. Virol. 46, 993-1002. Pierce J.H., and S.A. Aaronson. (1985). Myeloid cell transformation by ras-containing murlne sarcoma viruses. Mol. Cell. Biol. 5, 667-674. Pltot H.C. (1990). Carcinogenesis by chemicals: a multifaceted process. in The Cellular and Molecular Biology of Human Carcinogenesis. RK Boutwell, and LL Reigei. eds. (San Diego: Academic Press). pp. 81-109. Poirier Y., C. Kozak, and P. Joliculeur. (1988). Identification of a common helper provirus integration site in Abelson murlne leukemia virus induced lymphoma DNA. J. Virol. 62, 3985-3992. Ponder BAJ. (1990). inherited predisposition to cancer. Trends Genet. 6, 213-218. Potter M., and C. Boyce. (1962). induction of plasma cell neoplasms in strains Balb/c mice with mineral oil and mineral adiuvants. Nature 193, 1086-1087. Potter M.. and RC. MacCardie. (1964). Histology of developing plasma cell neoplasia induced by mineral oil in Baib/c mice. J. Natl. Cancer. institute 33, 497-515. 56 Potter M., J. Wax, E. Mushinski, S. Brust, M. Babonits, F. Wiener, J.F. Mushinski, D. Mezeblsh, R. Skuria, U. Rapp, and H.C. Morse ill. (1986). Rapid induction of plasmacytomas in mice by pristane. and a murlne recombinant retrovirus containing an avian v-myc and a defective raf oncogene. Curr. Topics. Micro. lmmunol. 132, 40-43. Preud’homme J.L., G. Fiandrin, M.T. Daniel, and JG Brouet. (1975). Burkitt’s tumor cells In acute leukemia. Blood 46, 990-992. Preud’homme J.L, K. Deilagi, P. Gugliemi, LB. Vogler, F. Danon, GM. Lenoir, F. Valensi, and JG Brouet. (1985). Immunologic markers of Burkitt’s lymphoma cells. in Burkitt’s lymphoma: a human cancer model. G.M. Lenoir, G. O’Conor, and C.L.M. Olweny. eds. (Lyon: lARC Scientific publication No. 60). pp. 47-64. Raghoebier S., J.H.J.M. van Krieken, J.C. KIuin-Neiemans, A. Gillis, G.J.B. van Ommen, AM. Ginsberg, M. Raffeid, and PM. Kluin. (1991). Oncogene rearrangements in chronic B-cell leukemia. Blood 77, 1560-1564. Rechavi G., D. Givoi, and E. Canaani. (1982). Activation of a cellular oncogene by DNA rearrangement: possible involvement of an lS-like element. Nature 300, 607-611. Reed J.C., M. Cuddy, T. Slabiak, C.M. Croce, and PC. Nowell. (1988). Oncogenlc potential of bcI-2 demonstrated by gene transfer. Nature 336, 259-261. Reed J.C., S. Hardar, C. M. Croce, and MP. Cuddy. (1990). Complementatlon by bcI-2 and c-Ha-ras oncogenes in malignant transformation of rat embryo fibroblasts. Mol. Cell. Biol. 10, 4370-4374. leokh R., Rouauit J.P. Wahbi K Gadoux M. and others. (1991). A chromosome 12 coding region is juxtaposed to the MYC protooncogene locus in a t(8; 12)(q24;q22) translocation in a case of B-cell chronic lymphocytic leukemia. Genes Chromosom. Cancer 3, 24-36. Scalenghe F., E. Turco. J.-E. Edstrom, V. Pirrotta, and M. Meili. (1981). Microdissection and cloning of DNA from specific region of Drosophila meianogaster polytene chromosomes. Chromosoma 82, 205-216. Schwab M., K. Aiitalo, K-H. Klempnauer, H.E. Varmus. J.M. Bishop, F. Gilbert, G. Brodeur, M. Golstein. and J. Trent. (1983). Amplified DNA with limited homology to myc cellular oncogene is shared by human neuroblastoma tumor cell lines and a neuroblastoma tumor. Nature 305, 245-248. Schwartz RC, and ON. Witte. (1988). The role of multiple oncogenes in hematopoietic neoplasia. Mutation Research 195, 245-253 . Schwartz R.C.. L.W. Stanton. S.C. Riley. K.B. Marcu, and ON. Witte. (1986a). Synergism of v-myc and v-Ha-ras in the in vitro neoplastic progression of murlne lymphoid cells. Mol. Cell. Biol. 6, 3221-3231. Seremetis S., G. lnghlraml, D. Ferraro, E.W. Newcomb, D.M. Knowles, G.-P. Dotto, and R. Dalia-Favera. (1989). Transforrnatlon and plasmacytoid differentiation of EBV-Infected human B lymphoblasts by ras oncogenes. Science 243. 660-663. Seth A., E. Priel, and Vande Woude GP. (1987). Nucleotide triphosphate—dependent DNA-binding properties of mos protein. Proc. Natl. Acad. Sci. USA 84, 3560-3564. Seto M., U. Jaeger, R.D. Hockett, W. Graninger, S. Bennett, P. Goldman, and SJ. Korsmeyer. (1988). Alternative promoters and axons. somatic mutation and deregulation of the bcI-2-lg fusion gene in lymphoma. EMBO J. 57 Shen-0ng G.L, E.J. Keath, S.P. Piccoli, and MD. Cole. (1982). Novel myc oncogene RNA from abortive immunoglobulin-gene recombination in mouse plasmacytomas. Cell 31, 443-452. Shen-Ong G.LC., M. Potter, J.F. Mushinski, S. Lavu, and E.P. Raddy. (1984). Activation of the myc locus hybridization Insertionai mutagenesis in piasmacytoid iymphosarcomas. Science 226, 1077-1080. Shepard J.S., D.H. Wurster-Hili. 0.S. Pettenglil, and GD. Sorenson. (1974a). Glemsa banded chromosomes of mouse myeloma in relationship to oncogeniclty. Cytogenet. Cell Genet. 13, 279-304. Shepard J.S., 0.6. Pettenglil, D.H. Wurster-Hiil, and GD. Sorenson. (1974b). Karyotype marker formation and oncogenicity in mouse plasmacytomas. J. Natl. Cancer inst. 56, 1003-1011. Shepard J.S., 0.6. Pettenglil, D.H. Wurster-Hill, and D. Sorenson . (1978). A specific chromosome breakpoint associated with mouse plasmacytomas. J. Natl. Cancer inst. 61, 225-256. Shih C., L Padhy, M. Murray, and RA. Weinberg. (1981). Transforming genes of carcinomas and neuroblastomas introduced into mouse fibroblasts. Nature 290, 260-264. Shih C.-C., J.P. Stoye. and J.M. Coffin . (1988). Highly preferred targets for retrovirus integration. Cell 53, 531-537. Shimizu K, M. Goldfarb, Y. Suard, M. Perucho, Y. Li, T. Kamata, J. Feramisco, E. Stavnezer, J. Fogh, and M.H. Wigler. (1983). Three human transforming genes are related to the viral ras oncogenes. Proc. Natl. Acad. Sci. USA 80. 2112-2116. Shimizu K, Y. Nakatsu, M. Seklguchl, K Hokamura, K Tanaka, M. Terada, and T. Sugimura. (1985). Molecular cloning of an activated human oncogene. homologous to wet. from primary stomach cancer. Proc. Natl. Acad. Sci. USA 82, 5641-5645. Siden E.J., D. Baltimore, D. Clark. and NE. Rosenberg. (1979). lmmunoglobulln synthesis by lymphoid cells transformed in vitro by Abelson murlne leukemia virus. Cell 16. 389-396. Silver J., and C. Kozak. (1986). Common proviral integration region on mouse chromosome 7 in lymphomas and myelogenous leukemias induced by Friend murlne leukemia virus. J. Virol. 57, 526-533. Skiar M.D., B.J. White, and WP. Rowe. (1974). Initiation of oncogenic transformation of mouse lymphocytes in vitro by Abelson Leukemia Vinis. Proc. Natl. Acad. Sci. USA 71, 4077-4081. Stehelin 0, RV. Guntaka, H.E. Varmus. and J.M. Bishop. (1976a). Purification of DNA complimentary to nucleotide sequences required for neoplastic transformation of fibroblasts by Avian sarcoma viruses. J. Mol. Biol. 101, 349-365. Stiles CD, and AA. Kawahara. (1978). The growth behavior of virus-transforrned cells in nude mice. in The Nude Mouse in Experimental and Clinical Research. J. Fogh, and B. Gioranella. eds. (New York: Academic Press). pp. 385-409. Stocking C., C. Loliger, M. Kawai, S. Suciu, N. Gough, and W. 0stertag. (1988). identification for genes involved in growth autonomy of hematopoietic cells by analysis of factor-independent mutants. Cell 53. 869-879. Suematsu S., T. Matsuda, K Aozasa, S. Aklra, N. Nakano, S. 0hno, J.-l. Mlyazakl, K-l. Yarnamura, T. Hirano, and T. Klshimoto. (1989). lgG 1 plasmacytosis in interleukin 6 transgenic mice. Proc. Natl. Acad. Sci. USA 86, 7547-7551. 58 Swendeman S., and DA. Thoriey-Lawson. (1987). The activation antigen BLAST-2, when shed, is an autocrine BCGF for normal and transformed B cells. EMBO J. 6, 1637-1642. Taira M., T. Yoshida, K. Miyagawak. and H. Sakamoto. M. Terada. and T. Sugimura. (1987). CDNA sequence of human transforming gene hst and identification of the coding sequence required for the transforming activity. Proc. Natl. Acad. Sci. USA 84. 2980-2984. Takada D., and T. Osato. (1979). Analysis of the transformation of human lymphocytes by Epstein-Barr Virus l. sequential occurrence from the virus-deterrnined nuclear antigen synthesis, to blastogenesis, to DNA synthesis. lntervirology 11, 30-39. Takahashi M., J. Ritz, and GM. Cooper. (1985). Activation of a novel human transforming gene, ret, by DNA rearrangement. Cell 42, 581-588. Tedder T.F., LT. Clement, and MD. COOper. (1984). Expression of C3d receptor during human B cell differentiation: lmmunofluorescence analysis with the HB5 monoclonal antibody. J. Immunol. 133. 668-673. Tsichlls P N., G. Strauss, and PH Liu. (1983a). A common region for proviral DNA integration in MoMuLV-induced rat thymic lymphomas. Nature 302, 445-449. Tsichlls P.N., P.G. Strauss. and CA. Kozak. (1984). Cellular DNA region involved in induction of thymic lymphoma(MIvi-z) maps to chromosome 15. Moi. Cell. Biol. 4, 997-1000. Tsichlls P.N., P.G. Strauss, and MA. Lohse. (19853). Concerted DNA rearrangements in Moloney murlne leukemia virus-induced thymomas: a potential synergistic relationship in oncogenesis. J. Virol. 56, 258-267. Tsujimoto Y. (1989). Overexpression of the human bcI-2 gene product results in growth enhancement of Epstein-Barr virus-immortalized B cells. Proc. Natl. Acad. Sci. USA 86, 1958-1962. Tsujimoto Y., J. Yunis, L 0norato-Showe, J. Erikson, P.C. Nowell, and CM. Croce. (1984a). Molecular cloning of the chromosomal breakpoint of B-ceii lymphomas and leukemias with the t(11;14) chromosomal translocation. Science 224, 1403-1406. Tsujimoto Y., LR. Finger, J. Yunis, P.C. Nowell, and CM. Croce. (1984b). Cloning of the chromosome breakpoint of neoplastic B cells with the t(14;18) chromosome translocation. Science 226, 1097-1099. Tsujimoto Y., E. Jaffe, J. Cossman, J. Gorham, P.C. Nowell. and CM. Croce. (1985a). Clustering of breakpoints on chromosome 11 in human B-cell neoplasms with the t(11;14) chromosome translocation. Nature 315, 340-343. Tsujimoto Y., J. Gorham, J. Cossman, E. Jaffe, and CM. Croce. (1985b). The t(14;18) chromosomal translocations involved in B-ceii neoplasms result from mistakes in VDJ joining. Science, 229 1390—1392. Tsujimoto Y., J. Cossman, E. Jaffe, and CM. Croce. (1985c). involvement of the bcI-2 gene in human follicular lymphoma. Science 228, 1440-1443. van der Berghe H., K Vermaelen, A. Louwagie, A. Criei, C. Mecuccl, and J.-P. Vaerman. (1984). High incidence of chromosome abnorrnallties In lgGa myeloma. Cancer Genet. Cytogenet. 11, 381-387. van Lohuizen M., M. Breuer, and A. Bems. (1989a). N-myc is frequently activated by proviral integration in MuLV induced T cell Lymphomas. EMBO J. 8. 133-136. 59 van Lohuizen M., S. Verbeek, S. Scheijen, E. Wientjens, H. van der Guiden, and A Bems. (1991). Cell 65, 737-752. identification of cooperating oncogenes in Eu-myc transgenic mice by provirus tagging. van Ness B., M. Shapiro, P.E. Kelley, R.P. Perry, M. Weigert, P. D’Eustachio, and F. Ruddle. Varmus H. (1989). A historical overview of oncogenes. ln Oncogenes and the Molecular Origins of Cancer. R.A. Weinberg ed. (New York: Cold Spring Harbor Laboratory Press). pp. 3-44. Vaux D.L, 8. Cory and J.M. Adams. (1988). Bel-2 gene promotes haemopoietlc cell survival and cooperates with c-myc to immortalize pre-B cells. Nature 335, 440-442. Vijaya S., D.L. Steffen, C. Kozak, and H.L Robinson. (1987). A region with frequent proviral insertions in Moloney murine leukemia virus induced rat thymomas. J. Virol. 61, 1164-1170. Villemur R., Y. Monczak, E. Rassart, C. Kozak, and P. Jolicoeur. (1987). identification of a new common proviral integration site in cross passage a murlne leukemia virus-induced mouse thymoma DNA. Mol. Cell. Biol. 7, 512-522. Vlnk A, P. Coulie, G. Warnler, J.-C. Renauld, M. Stevens, D. Donckers, and J. Van Snick . (1990). Mouse plasmacytoma growth in vivo: enhancement by interleukin 6 (lL-6) and inhibition by antibodies directed against iL-6 or its receptor. J. Exp. Med. 172, 997-1000. Voisky D.J., T. Gross, F. Sinangil, C. Kuszynski, R. Bartzatt, T. Dambaugh, and E. Kieff. (1984). Expression of Epstein-Barr Virus(EBV) DNA and cloned DNA fragments in human lymphocytes following Sendai virus envelop-mediated gene transfer. Proc. Natl. Acad. Sci. USA 81, 5926-5930. Wang D., D. Liebowitz, and E. Kieffe. (1985). The EBV membrane protein expressed in immortalized lymphocytes transforms established rodent cells. Cell 43, 831-840. Wang D., D. Liebowitz, F. Wang, 0. Gregory, A. Rickinson, R. Larson, T. Springer, and E. Kieff. (1988). Epstein-Barr virus latent infection membrane protein alters the human B-lymphocyte phenotype: deletion of the amino terminus abolishes activity. J. Virol. 62, 4173-4184. Wang F., OD. Gregory, 0. Sample, M. Rowe, D. Liebowitz, R. Murray, A. Rickinson, and E. Kieff. (1990). Epstein-Barr virus latent membrane protein (LMP1) and nuclear protein 2 and 3c are effectors of phenotypic changes in B lymphocytes: EBNA-2 and LMP1 cooperatively induce CD23. J. Virol. 64, 2309-2328. Whitlock CA. and 0N. Witte. (1981). Abelson virus-infected cells can exhibit restricted in vitro growth and low oncogenic potential. J. Virol. 40. 577-584. Whitlock C.A., S.F. Ziegler, and 0N. Witte. (1983b). Progression of the transformed phenotype in clonal lines of Abelson virus-infected lymphocytes. Mol. Cell. Biol. 3, 596-604. Ymer S., W.Q.J. Tucker, C.J. Sanderson, A.J. Hapel, H.D. Campbell, and LG. Young. (1985). Constitutive synthesis of interleukin-3 by leukemia cell line WEHi-3B is due to retrovlral insertion near the gene. Nature 317, 255-259. Yosida T.H., H.T. lmai, and U. Moriwaki. (1970). Chromosomal alteration and development of tumors XXI cytogenetic studies of primary plasma cell neoplasms induced in Balb/c mice. J. Natl. Cancer Inst. 45, 411-418. Young D., G. Waitches, Birchmeier, 0. Fasano, and M. Wigler. (1986). isolation and characterization of a new cellular oncogene encoding a protein with multiple potential transmembrane domains. Cell 45, 711-719. Youm; DGCE’S Yunis Yunis Zach E in bio: Canoe zur H; 60 Young J.C., C. Mikttail, B. Gishizky, and 0N. Witte. (1991). Hyperexpresslon of interleukin-7 is not necessary or sufficient for transformation of a pre-B lymphoid cell line. Mol. Cell. Biol. 11, 854-864. Yunis J.J. (1976). High resolution of human chromosomes. Science 191, 1268-1270. Yunis J.J. (1983). The chromosomal basis of human neoplasia. Science 221, 227-236. Zech L, U. Haglund, K Nilsson, and G. Klein. (1976). Characteristics chromosomal abnormalities in biopsies and lymphoid cell lines from patients with Burkitt and non-Burkitt lymphomas. int. J. Cancer 17, 47-56. zur Hausen H. (1980). Oncogenlc herpesviruses. in DNA Tumor Virus. J. Tooze ed. (New Work: Cold Spring Harbor Laboratory). pp. 755-757. Chapter 2 Vol. 178, No. 3, 1991 BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS August15,1991 Tumorigenesis of a V—Ha-gas-Bxpressing Pre-B Cell Line Selects for c-ugg Activation Shu-Chih Chen, Diane Redenius, and Richard C. Schwartz Department of Microbiology, Michigan State University, East Lansing, MI 48824-1101 Received June 24, 1991 Seven tumors independently derived from a v-Ha-ggg-expressing pre-B cell line were examined to determine the oncogene activations cooperating with v-Ha-gag in 13 vivo tumor progression. The pre-B cell line was generated by infection with Moloney murine leukemia virus (MoMuLV) and a MoMuLV-derived recombinant expressing v-Ha-ggg. Two of seven tumors possessed a MoMuLV integration immediately upstream and in reverse transcriptional orientation to c-m. This correlated with a 3-fold increased level of c-myg mRNA. Two other tumors displayed elevated c-myg mRNA levels, although the mechanism of enhanced expression was unclear. Thus the tumor progression of a v-Ha-gag- expressing murine pre-B cell line selects for the activation of c-myg. elem Academic Press . Inc . It is generally accepted that tumorigenesis proceeds through multiple events involving the altered expression or structure of specific oncogenes that function in the regulation of cellular growth. The requirement of two cooperating' oncogenes for the 13_ giggg neoplastic transformation of many primary cells may be a reflection of this process. Several examples of oncogene cooperativity in lymphoid transformation have been described in experimental systems. Infection by Abelson murine leukemia virus accelerates the induction of plasmacytomas by pristane (1). These tumors not only express v-g_l, but possess the same c-myg translocations observed in tumors induced by pristane alone. Cooperativity between v-ggg and v-myg has been observed in the induction of both 3 lymphomas (2) and plasmacytomas (3). A synergy of v-Ha-gag and v-myg has been observed in the i3 giggg transformation of pre-B lymphoid cells (4). Either v-Ha-ggg or v-ggg can cooperate with activated my; in the transformation of Ep-myg transgenic pre-B lymphoid cells (5,6). While in 315:9 manipulations have revealed pairs of oncogenes capable of eliciting full neoplastic transformation, there have been few tests of what oncogenes might be selected 13 yigg to achieve transformation given the prior expression of a single oncogene that is incapable by itself of eliciting transformation. The retroviral activation of c-fms has been observed in a monocytic tumor 61 62 Vol. 178, No. 3. 1991 BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS induced by a c-myg-expressing retrovirus (7). 3;; mutations were observed in some lymphomas generated in Ep-myg transgenic mice (8). In earlier studies, we found that v-Ha-m—expressing pre-B lymphoid cells displayed an intermediate transformed phenotype and were infrequently tumorigenic (4). The irregular occurrence of tumors derived from these cells suggested continued neoplastic progression in gigg, or selection in zigg for pro-existing subpopulatlons containing additional neoplastic mutations. Since v-myg was found to cooperate with v-Ha-ggg in the in giggg transformation of pre-B lymphoid cells, we decided to investigate the status of c-myg in tumors derived from a v-Ha-gag-expressing pre-B cell line. IAIBRIALS AND METHODS W. R2, a v-Ha-m-transformed murine pre-B cell line, and tumors derived from R2 are described in Schwartz et al. (4). Tumor cell lines were produced from explanted tumors by dispersal onto feeder cultures of adherent bone marrow cells (9). Cell lines were cultured over feeder cells in RPMI 1640 with St fetal calf serum and 5 x 10-5 H 2-mercaptoethanol. Nucleic acid analyses. Cytoplasmic RNA was isolated by a sodium dodecyl sulfate-urea procedure as described by Schwartz et al. (10). Poly A+ RNA was selected by oligo-dT cellulose chromatography. RNA was denatured, electrophoresed in a formaldehyde-1t agarose gel and transferred to Nytran (Schleicher and Schuell). High molecular weight DNA was isolated from nuclei collected in the preceding RNA isolation procedure as described in Schwartz et al. (4). DNA was digested with restriction enzymes as noted, electrophoresed through agarose and transferred to Nytran. Hybridization probes were prepared by nick translation through the incorporation of [a-32P1dATP (3000 Ci/mmol; ICN). The v-Ha-ggg probe was a 0.46 kb EcoRI fragment corresponding to v-Ha-gag encoding sequences (11). The en! probe was a 0.8 kb BamHI fragment from the gag region of Friend murine leukemia virus (12). The c-myg probes were the 4.7 kb HindIII fragment of murine c-myg encompassing exons 1, 2 and 3 (Figures 3 and 6) and the 0.8 kb SmaI-Sac]: fragment encompassing S' upstream flanking sequences and part of exon 1 (Figures 4 and 5). The rat glyceraldehyde-3-phosphate dehydrogenase (rGAPDH) probe was a cloned 1.3 kb cDNA (13). All hybridizations were performed under aqueous conditions in 5 x SSC at 65°C and washed to a stringency of 0.1 x SSC at 65°C. RESULTS Seven tumors independently derived from a v-Ha-ggg-expressing pro-8 cell line were studied in regard to their retroviral integration sites and status of c-mxg structure and expression. These tumors were derived from the clonal R2 cell line that was generated by infection of fresh murine bone marrow with a recombinant v-Ha-ggg-expressing retrovirus and MoMuLV (4). This cell line is dependent upon a bone marrow-derived cellular feeder layer for growth and gives rise to tumors infrequently. r a e v d fro . In order to verify that the tumors were derived from R2, sites of integration of the vbfla-ras-expressing retrovirus were compared between that cell line and the tumors. Southern hybridization of BcoRI-digested DNA with a v—Ha-zag probe showed that the tumors contained 63 Vol. 178, No. 3, 1991 BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS FIGURI 1. Viral :3; integrations. Southern blot analysis of DNAs from liver (L), R2, and seven tumors (1-7). DNA was digested with lcoRI and lOpg of each sample was electrophoresed through O.BI agarose. The blot was probed for v-Ha-ggg. Size markers are the positions of an ethidium bromide-stained Hind III digest of bacteriophage l and are denoted in kilobases. the same 5.3 kb proviral integration fragment as R2 (Figure 1). In addition to the 5.3 kb fragment, there is a 23 kb fragment representing the endogenous c-Ha-ggg gene in all the DNAs, and an additional 4.2 kb fragment in tumor 7. This 4.2 kb fragment represents an additional v-Ha-zgg proviral insertion site (data not shown). a v v s . If the tumors resulted from genetic lesions subsequent to the introduction of v-Ra;;gg, then they would be expected to be clonal outgrowths from the parental R2 cell line. R2 was generated by infection with a recombinant v-Ha-gag—expressing retrovirus and MoMuLV. since proviral integrations can occur subsequent to the original integration event, it is reasonable to expect clonal outgrowths to contain unique sites of proviral integration in addition to any common sites derived from the parent. Examination of integrations of the v-Ha-ggg retrovirus revealed only one new site in tumor 7 (Figure 1). We then examined the sites of MoMuLV integration in R2 and the tumors by Southern hybridization of BglII- digested DNA, using a probe for the ecotropic MuLV egg gene. All of the tumors possessed gag-hybridizing BglII fragments unique to the tumors and not present in R2 (Figure 2). Conversely, some fragments present in R2 were absent in the tumor cell lines. Fragments unique to the tumors represent either new proviral integrations or rearrangements of pre-existing integrations. The tumors (with the exception of tumor 4) clearly possess guy-hybridizing BglII fragments more similar to each other than to R2, although individual tumors also possess unique BglII fragments. These data suggest that the tumors (with the exception of tumor 4) are clonal outgrowths derived from a single subclone of R2. Tumor 4 appears more closely related to the original R2 isolate. 64 Vol. 178, No. 3, 1991 BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS fix its” ~ “istflyr‘i’lfis goals 2:. MoMuLV integrations. Southern blot analysis of blue from BALD/c mouse liver (1.), R2, and seven tumors (1-7). DNA was digested with lglII and lOug of each sample was electrophoresed through 0.8! agarose. The blot was probed for murlne ecotropic m sequences. Size markers are the positions of an ethidium bromide-stained Hindu! digest of bacteriophage .\ and are denoted in kilobases. Rearranganent of cm in tumors 1 and 3. Southern blot analysis of Blue from liver (L), R2 and seven tumors (1-7). DNA was digested with Icon: and long of each sample was eletrophoresed through 0.3\ agarose. Size markers to the left of photographs are the positions of an ethidium bromide- stained flindIII digest of bacteriophage l and are denoted in kilobases. Raving demonstrated that v-m can cooperate with v-aa-m in the in mm neoplastic progression of pre-B lymphoid cells (4), we investigated the status of the c-m locus in the tumors. Southern hybridization of lcoRI-digested blue with a c-m probe revealed that tumors 1 and 3 possessed a m- hybridizing fragment of mid: in addition to the normal 23kb fragment (Fig. 3). Since the tumors displayed several proviral integrations, we investigated whether the rearrangement of c-m in tumors 1 and 3 was caused by integration of MoMuLV. Assuming a proviral integration site near cm, and with complete restriction maps of both MoMuLV and cm, we could' predict the sizes of restriction fragments that would be revealed by a c-m probe in a Southern analysis (Fig. 4). Assuming that an intact MoMuLV provirus had integrated adjacent to exon 1 of c-m in a reverse transcriptional orientation (the 5' long terminal repeat (LTR) juxtaposed to exon 1), a series of expected sizes were calculated (Table 1). We performed double digestions with xbaI/xhoI, SacI/xhoI and XpnI/XhoI to examine the presence of a MoMuLV LTR pattern of restriction sites in the vicinity of c-m (Fig. 4). The data generated by digestions with restriction enzymes specific for LTR sites were in excellent agreement with the predicted sizes (Fig. 4 and Table 1). These data suggested integration of a MoMuLV genome within lOObp of the 5'-terminue of c-m. In order to further verify the presence of KoHuLV near c-m in tumors 1 and 3, BglII-digested DNA was analyzed by Southern blot with a probe for the MuLV m gene and a probe for c-m that included exon 1 and 5' 65 Vol. 178, No. 3, 1991 BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS XbaI—Xhol SacI-Xhol Kpnl-Xho as I a a5 1‘ a 'sa' f ’f? 23. - I 3'3: . .‘- t as -. 4L4. ‘ ‘_ .. .. K -——3.7 - 2. ' 9' :13: .- ‘No . s w- +——t9 ‘.4 #1.? -~ o——4190 ‘ s——1Lss (176 2..— L P sum '"“'5 w a. i 1 i I ITII'I 1 1 fit. It I = H» wit a a JIL L I 9 A. p g n g g. rxgggg 4. MoMuLV integration near c-myc. Southern blot analysis of DNAs from R2 and tumors 1 and 3. DNA was digested and lOug of each sample was electrophoresed through 1.6t agarose. Size markers to the left are the positions of an ethidium bromide-stained BindIII digest of bacteriophage A and are denoted in kilobases. The positions of rearranged c-myg fragments are marked to the right of photographs and denoted in kilobases. DNA was doubly digested with either XbaI and XhoI, or SacI and XhoI, or anI and XhoI. The blot was probed with a SmaI-SacI fragment encompassing' upstream flanking sequences and part of the first exon of murine c-m. A schematic of the murlne c-myg locus with MoMuLV integrated upstream in a reverse transcriptional orientation is included. The following restriction sites are marked: KpnI (K), SacI (S), XbaI (Xb), SmaI (Sm), BclI (8c), AatII (A), NotI (N), XhoI (Xh) and 89111 (89). The probe is delineated by a line labeled '9' above the map. Note that probe ”P“ is interrupted by MoMuLV integration. flanking sequences extending to a SmaI site about SOObp upstream of c-m (Fig. 5). Tumors l and 3 displayed two identical rearranged c-myg-hybridizing fragments, one of which (see arrowheads) co-hybridized with the envelope Table 1. Restriction fragments in the 5' c-myg region of tumors 1 and 3 Fragment Rxpected Size Observed Size lxon l/LTR: XhoI-XDaI 809bp 750bp SacI-SacI 919 840 XhoI-anI 992 900 Upstream Region/LTR: XbaI-XbaI 1797 1700 SacI-SacI 1954 1870 xpnI-xpnt 4000‘ 3700 ‘ Estimated value since the region between the upstream KpnI site and c-myc is not completely sequenced. 66 Vol. 178, No. 3, 1991 BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS guises c-mr wuwzauuasu a: ”1, 2 a 4 5 _07 7 .« ' C-MVC ..' ‘-._.“ 'DIII ..III—sQ’ <::> GAFDN an. pl. 1.. ‘l’ II. 'II' J FIGURE 5. En! and c-mxg co-hybridize. southern blot analyses of DNAs from liver (L), R2 and tumors 1, 2, 3 and 5. DNA was digested with BglII and lOug of each sample was electrophoresed through 0.8t agarose. Size markers are the positions of an ethidium bromide-stained HindIII digest of bacteriophage A and are denoted in kilobases. Arrowheads mark co-hybridization. (A) The blot was probed for murlne ecotropic virus guy sequences. (3) The blot was probed for c-myg with a SmaI-SacI fragment encompassing upstream flanking sequences and part of exon 1. FIGURE 6. Elevated c-myg mRNA levels. Northern blot analyses of polyA+ RNA from R2 and seven tumor cell lines (1—7). Each sample of RNA was the polyA+ fraction selected from lSOug of total cytoplasmic RNA. The blot was probed for both c-myg and rGAPDH. Relative levels of c-myg mRNA were determined by densitometry and normalized to rGAPDH. region probe. An LTR probe detected fragments that co-migrate with both rearranged c-mJLc-hybridizing fragments indicating integration of both viral LTRs (data not shown). Clearly, tumors 1 and 3 had suffered MoMuLV integration in close proximity to c—myg. MoMuLV ' “on causes i ‘ 'ann of c-mvc. Having observed proviral insertion near c-mg in two tumors, we decided to test whether expression of c-myg mRNA was altered in these or any of the other tumors. Cytoplasmic poly A+ RNAs of R2 and the seven tumor cell lines were prepared from actively growing cells and examined by Northern hybridization (Fig. 6). The same blot was hybridized successively with c-M and rGAPDH probes. Hybridization to the rGAPDH probe provided a control for gel loading. Densitometry revealed that tumors 1 and 3 had 3—fold elevated levels of c-m mRNA. Tumors 6 and 7, which displayed no gross abnormalities in c-m, had 4- fold elevated levels of c-m mRNA. The c-M mRNAs were all 2.4kb in length, suggesting normal promoter usage and an unaltered RNA structure. DISCUSSION The data presented here demonstrate selection for c-M activation in the lymphomagenesis of a v-Ha-m—transformed murine pre-B lymphoid cell line. Four of seven tumors examined had 3 to 4—fold elevated levels of c—m mRNA. In the case of tumors 1 and 3, MoMuLV had integrated immediately 5' to c-m VoLW in a natu {330 othe (22) I'll 1m in 1 I!!! V-Ha 0th ever Vit! 67 Vol. 178. No. 3, 1991 BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS in a reverse transcriptional orientation, suggesting enhancer activation. The nature of the events leading to elevated levels of c-myg mRNA in the other two tumors is unclear. The activation. of c-myg, by retroviral integration is 'the prominent feature of lymphomas induced by avian leukosis virus (14,15). MoMuLV activation of c-myg is also a frequent event in T lymphomas induced by that virus (16,17). This report of MoMuLV activation of c-myg in a murine B lymphoma is novel. The structure of the MoMuLV integration in tumors 1 and 3 is reminiscent of retroviral integrations observed in murine T lymphomas (16- 19). The provirus is located upstream of c-myg in a reverse transcriptional orientation. The modest 3-fold increase in the steady state level of c-myg mRNA is similar to that observed by others in T lymphomas (18,20,21). We have examined v-Ha-m-induced B lymphomas for rearrangements at other frequent MoMuLV integration sites found in T lymphomas. Neither ulyi;1 (22), 513i;z (23), fllvi-B (24), Mlvi-4 (25), nor pim;1 (26) were found to be rearranged (data not shown). We have not yet examined a large enough panel of tumors to assess the importance of these frequent integration sites in B cell lymphomagenesis, but examination of MoMuLV integration sites may be fruitful in this system. The seven tumors studied possess MoMuLV integration-related restriction fragments unique to the tumors and absent from 32, the parental v-Ha-m-expressing cell line. This indicates that the tumors are clonal outgrowths presumably having advanced in tumor progression due to mutagenic events. The patterns of viral integration in six of the tumors are more similar to each other than to R2. This suggests that a single subclone of R2 was predisposed toward tumorigenesis. The presence of MoMuLV integration near c-myg in two of these six tumors indicates that retroviral activation of c-myg is at least the third event in the progression of these tumors. This suggests that the activation of oncogenes other than gag and my; may be important for tumorigenesis. Our finding of common MoMuLV integration sites in addition to two instances of integration near c-myg presents the possibility that sites of MoMuLV integration may identify additional genetic elements that can cooperate with v-Ha-gag in the tumor progression of B lymphoid cells. Acknowledgment: This work was supported by grant CA45360 from the National Cancer Institute. REFERENCES 1. Ohno, s., Higata, 5., Weiner, P., Babonits, N., Klein, 6., flushinski, J. 1., and Potter, H. (1984) J. Exp. Med. 159:1762-1777. 2. Rapp, U. R., Cleveland, J. L., Fredrickson, T. N., Holmes, K. L., Morse III, R. C., Jansen, H. w., Patschinsky T., and Sister, x. (1985) J. Virol. 55:23-33. 3. Troppmair, J., Potter, M., wax, J. 8., and Rapp, U. R. (1989) Proc. Natl. Acad. Sci. USA 86:9941-9945. 68 Vol. 178, No. 3, 1991 BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS 4. Schwartz, R. C., Stanton, L. 10., Riley, 8. C., Marcu, K. B., and Witte, O. N. (1986) Mol. Cell. Biol. 633221-3231. 5. Alexander, W. 8., Adams, J. N., and Cory, 8. (1989) hol. Cell. Biol. 9:67-73. 6. Langdon, W. Y., Harris, A. N., and Cory, S. (1989) Oncogene Res. ' :253-258. 7. Baumbach, N. R., Colston,B. N., and Cole, H. D. (1989) J. Virol. 62:3151-3155. 8. Alexander, W. 8., Bernard, 0., Cory, 8., and Adams, J. N. (1989) Oncogene 4:575-581. 9. Whitlock, C. A., Ziegler, S. F., Treiman, L. J., Stafford, J. 1., and Witte, O. N. (1983) Cell 32:903-911. 10. Schwartz, R. C., Sonenshein, C. 3., Bothwell, A. and Gefter, N. L. (1981) J. Immunol. 126:2104-2108. 11. Ellis, R. N., De Feo, D., Maryak, J. 14., Young, H. A., Shih, T. Y., Chang, 8. H., Lowy, D. R., and Scolnick, E. H. (1980) J. Virol. 36:408-420. 12. Silver, J., and Kozak, C. (1986) J. Virol. 57:526-533. 13. Port, P., Marty, L., Piechaczyk, N., 31 Salrouty, 8., Dani, C., Jeanteur, J., and Blanchard, J. H. (1985) Nucl. Acids Res. 13:1431- 1442. 14. Hayward, W. 8., Neel, B. C., and Astrin, S. N. (1981) Nature 290:475- 480. 15. Payne, G. 8., Bishop, J. N., and Varmus, H. E. (1982) Nature 295:209- 214. 16. Selten, C., Cuypers, H. T., Zylstra, N., Hilief, C. and Berns, A. (1984) EHBO J. 3:3215-3222. l7. Steffen, D. L. (1984) Proc. Natl. Acad. Sci. USA 81:2097-2101. 18. Corcoran, L. N., Adams, J. N., Dunn, A. P., and Cory, S. (1984) Cell 37:113-122. 19. Li, Y., Holland, C. A., Hartley, J. H. and Hopkins, N. (1984) Proc. Natl. Acad. Sci. USA 81:6806-6811. 20. Reicin, A., Yang, J. 9., Marcu, K. B., Fleissner, R., Xoehne, C. P. and O'Donnell. P. V. (1986) Mol. Cell. Biol. 6:4088-4092. 21. Steffen, D. L., and Nacar, E. Q. (1988) Virology 164:55-63. 22. Tsichlis, P. N., Strauss, P. C., and flu, L. F. (1983) Nature 302:445- 449. 23. Tsichlis, P. N., Strauss, P. C., and Lohse, H. A. (1985) .J. Virol. 56:258-267. 24. Tsichlis, P. N., Lohse, N. A., Szpirer, C., Szpirer, J., and Levan, G. (1985) J. Virol. 56:932-942. 25. Lazo, P. A., Lee, J. 8., and Tsichlis, P. N. (1990) Proc. Natl. Acad. Sci. USA 87:170-173. 26. Selten, C., Cuypers, H. T., and Berna, A. (1985) EMBO J. 4:1793-1798. Chapter 3 Cloning and characterization of a viral flanking region common to B lymphoid tumors derived from a v-Ha-ras infected pre-B cell line Shu-Chih Chen,1 Marge Strobel? Neal Copeland,2 Nancy Jenkins,2 and Richard C. Schwartz‘ lDepartment of Microbiology, Michigan State University, East Lansing, MI 48824-1 101 2Mammalian Genetics Laboratory, BRI-Basic Research Program, NCl-Frederick Cancer Research Facility, Frederick, Maryland 21 701 69 70 Abstract: A viral flanking region, designated as bone-1, common to B lymphoid tumors derived from a v-Ha-ras oncogene-transformed cell line, R2 (Chen et al., 1991) was cloned and characterized. This locus may be associated with the growth advantage of tumors derived from the R2 cell line. We were unable to detect any transcriptional activity over a 20 kb region in the vicinity of the bonc-1 locus. However, sequence homology within bonc—1 was found to the genome of other mammals, and chicken. The high evolutionary conservation within the bone-1 region and the presence of several open reading frames suggest that one or more sequences of function importance exist therein. Bone-1 was mapped to mouse chromosome 19, closely linked to the Ipr locus, and distinct from another frequent viral integration site, gin-1, on the same chromosome. The Ipr locus is linked to a recessive lymphoproliferation trait. Whether bonc-l is identical to Ipr awaits further investigation. The fact that it is close to the lpr locus provides a use for this locus in the search for genes responsible for the Ipr lymphoproliferation trait. 71 Introduction: Oncogenlc transformation by nonacute transforming retroviruses such as avian leukosis virus (ALV), mouse mammary tumor virus (MMTV), and murlne leukemia virus (MLV) depends on the activation of cellular protooncogenes by viral integration. Studies of frequent viral integration sites in animal tumors have been fruitful both in identifying new gene loci that could be involved in tumorigenesis, and in reconfirming the transformation potential of known protooncogenes (for review see Nusse, 1986a). Examples of the first case are pim-l (Cuypers et al., 1984), int-2, (Nusse and Varmus, 1982) and int-2 (Dickson et al., 1984). Pim-1 was identified as a common region of insertion for mink cell focus-forming (MCF) virus or other MuLV in T cell lymphomas of several mouse strains (Cuypers et al., 1984; Selten et al., 1985). Pim-1 encodes a serine kinase (Selten et al., 1986), and may thus participate in cell signaling. Studies of transgenic mice carrying the pim- 1 gene have verified its role in T lymphoid neoplasia (van Lohuizen et al., 1989b; Breuer et al., 1989a; Breuer et al., 1991). Increased expression of int-1 and int-2 as a result of MMTV insertion is thought to contribute to the formation of mouse mammary tumors through an autocrine loop (Aaronson, 1991). Both int-1 and int- 2 are expressed temporally in a very restricted pattern during early development of the mouse, and the protein products show the characteristics of genes encoding secreted factors (for review see Nusse, 1988). Studies of transgenic mice bearing activated int-1 (T sukamoto et al., 1988) or int-2 (Muller et al., 1990) confirm their oncogenic properties. Such animals develop mammary hyperplasia, and those with an int-1 transgene gradually develop mammary carcinomas with age. A wide range of cellular oncogenes activated by viral integration has been 72 found in a variety of tumors and these integration events have been implicated in the malignant phenotypes of the tumors. These include lL-3 (Morishita et al., 1988), GM-CSF (Stocking et al., 1988), CSF-1 (Baumbach et al., 1988), c-Ha-ras (lhle et al., 1989), c-erb B (Fung et al., 1983), c-myb (Shen-Ong et al., 1984), N- myc ( van Lohuizen et al., 1989a), p53 (Hicks and Mowat, 1988) and, most frequently, c-myc (Hayward et al., 1981; Neel et al., 1981; Payne et al., 1982; Westaway et al., 1984; Swift et al., 1985; Corcoran et al., 1984; Selten et al, 1984; Li et al., 1984; Steffen, 1984; O’Donnell et al., 1985; Neil et al., 1984). Here we describe a locus that we have designated as bone-1. Bonc-1 is one of several common integration sites found in 6 of 7 Balb/c B lymphoid tumor cell lines (Chen et al., 1991) (Chapter 2). These 7 tumor cell lines were derived from independent tumors which were collected after tumor challenge of Balb/c mice with a cell line (R2) established after infecting Balb/c bone marrow cells with a retroviral vector carrying the v-Ha-ras oncogene and helper Moloney murine leukemia virus (MoMuLV) (Schwartz et al., 1986a). The long latency and infrequent occurrence of tumor development within these animals led us to hypothesize the involvement of secondary events in the tumorigenesis of these 7 cell lines (Chen et al., 1991). The association of gene activation through viral integration to these secondary events was suggested by the observation of common viral integration sites in 6 of these 7 tumor cell lines (Chen et al., 1991). This result indicates the outgrth of a transformed subclone from the original R2 cell population during the period of tumor progression. Studies of the viral flanking regions may reveal the molecular basis of the growth advantage possessed by this subclone, and, furthermore, the putative roles of the flanking regions in oncogenesis of B lymphoid 73 tumors. Materials and Methods: Mice. Balb/c.lpr mice were a gift from The Jackson Laboratories (Ban Harbor, Maine). Cell culture. R2 and tumor cell lines were maintained on cellular feeder cultures as described in Chen et al. (1991). Only short term cultures of two weeks or less were used for molecular analyses to avoid accumulation of genetic alterations in vitro. Nucleic acid isolation. High-molecular-weight DNA was isolated from nuclei collected as described by Schwartz et al. (1986a). Cytoplasmic RNA was isolated by a sodium dodecyl sulfate (SDS)-urea procedure as described by Schwartz et al. (1981). Poly A" RNA was selected by oligo-dT cellulose chromatography (Aviv and Leder. 1975). Testicle RNA was prepared by a LiCl/urea method with a motor driven polytron homogenizer (Auffray and Rougeon, 1980). Molecular cloning. DNA from tumor 5 (T 5) was partially digested with Sau3A and partially end-filled with dGTP and dATP, and then cloned into a Xhol half-site LambdaGEM-11 vector (Promega). The ligated DNA was packaged using a Packagene kit(Promega). The packaged DNA was titered and plated onto LB plates using LE392 (Promega) as host. The library was screened by a env or LTR enhancer-specific probe as described (Chen et al., 1991). Positive clones were purified and used to prepare phage DNA (Sambrook et al., 1989). Viral flanking regions were identified by hybridization and restriction mapping analyses. DNA fragments which contain viral flanking regions and with appropriate restriction site 74 ends were then subcloned into pBIuescript IlKS+ (Stratagene). A Pstl DNA fragment of bone-1 was used to "walk" through a NIH3T3 phage genomic library (a gift from Dr. DeWitt at Michigan State University). This library was generated in Lambda FixTM ll (Stratagene). Clones 11 and 32 derived from this library were isolated by similar approaches to those described above. Three SacI DNA fragments of clone 11 and clone 32, respectively, were also subcloned into the pBIuescript KSII + vector for fine structure restriction mapping. Sequences unique to clone 11 and clone 32 were identified by hybridization of DNA blots used in the restriction mapping with a labelled liver DNA probe. Since 1/3 of total genomic DNA consists of repetitive sequences, restriction fragments that hybridize strongly to labelled liver DNA contain repetitive sequences unlikely to encode gene products. Therefore, sequences of these restriction fragments were excluded in northern analyses for detecting mRNA expression of bonc-1. Sequences unique to bone-1 included the retire insert fragment of clone 11 except the two Xbal fragments in the very ends of the insert fragment, and included the 2.2 kb SacI fragment in the right side of the clone 32 insert fragment as shown in Figure 2. Blots and Hybridization analyses. Southern and Northern blot hybridization procedures were performed as described by Chen et al. (1991) except that a oligomer random priming kit (USB) was occasionally used as the probe labelling method. Plaque lifts were prepared as described in Sambrook et al. (1989). All washing conditions were performed to a stringency of 0.1 x SSPE at 65° C except in the case ofthe zoo blot where conditions are indicated in the figure legend. The rat gcheraldehyde-S-phosphate dehydrogenase (rGAPDH) and env probes were as described (Chen et al., 1991). The 03P2 probe was a 1.3 kb Pstl fragment 75 from the bone-1 locus (this paper). Restriction mapping. Restriction enzyme sites in the multiple cloning regions of the various vectors were used as reference points for mapping. Double digestion with these enzymes and sites in the cloned DNA were performed to construct the restriction map of bonc-l. DNA sequenclng. The 03P2 fragment was subcloned into Mp19 for single-strand- sequencing. Subclones which produced either (+) or (-) strand of the recombinant M13 phage DNAa were obtained. DNA sequences were determined by the dideoxy sequencing procedure (Sanger et al., 1977) using a sequencing kit from USB. The predicted sequence was analyzed with the sequence analysis software package of the University of Wisconsin Genetics Computer Group (Devereux et al., 1986) or Genepro4.2 (Riverside Scientific, Seattle). 76 Results: Cloning of a tumor specific viral flanking region: bonc-1 A genomic library, lambda T5, was constructed from DNA of tumor cell line 5 (T 5) to isolate tumor specific viral flanking regions. T5 was chosen because it apparently contains the highest number of viral integration sites as judged by the number of hybridized fragments on a southern blot hybridization analysis with a virus-specific probe for the envelope gene of ecotropic viruses (Chen et al., 1991). Four positive clones (A-D) were originally identified from 2 x 105 recombinant phages. Viral flanking regions of these clones were subcloned by first identifying the integration junction fragment and then isolating the non-viral subfragment from the junction fragment. Subsequent analyses of viral flanking sequences of individual clones revealed that clones A and B were derived from a viral integration site that was already present in the R2 parental cell line (data not shown); clone D consisted of the viral flanking region of an endogenous virus (data not shown); and clone C contained the flanking region of a tumor-specific virus integration site (Figure 1). Southern blot hybridization of EcoRl-digested DNA with a probe derived from the flanking region of clone C (CSP2) demonstrated that six of the seven tumor cell lines (lanes 3, 4, 5, 7, 8, and 9) contained a 20 kb fragment in addition to the 11 kb germ-line fragment, whereas the R2 cell line and the tumor 4 (T 4) contained only the same germ-line fragment (lanes 2 and 6) as that in liver (lane 1). The size difference (9 kb) of these two fragments was about the size of a MoMuLV provirus (8.8 kb). Since EcoRI does not out within the MoMuLV sequence, the 20 kb fragment probably resulted from an integration of an intact virus. This locus was designated as bone-1, a putative B cell oncogene. 77 123456789 23> . .. 9.4. w ‘ " a ‘1‘? H ‘. ~4- ez.-.a ‘N g. 44' Figure 1. Tumor specific viral integration in the bonc-1 locus. Southern blot analysis of DNAs from liver (lane 1), R2 (lane 2) and tumor cells (lanes 3—9). DNA was digested with EcoRI and 10 pg of each sample was electrophoresed through 0.8 % agarose. The blot was probed for bone-1. Size markers are the positions of an ethidium bromide-stained Hindlll digest of bacteriophage l, and are denoted in kilobases. Expr from 401 expressi kb C3P£ Sinc activate we dec walking. rescree! since Ni donesl clone 3: revealec 20.5 kb side on and Clo restrict“ Schlon in FigUr. Nor 20.5 kb 099(31in 78 Expression of bonc-1 was examined by northern blots of poly A” RNA isolated from 400 micrograms of cytoplasmic RNA to ensure the detection of low levels of expression. No mRNA was detectable in R2 or the tumor cell lines using the 1.3 kb CSP2 fragment as a probe (data not shown). Since the viral LTR may function over a relatively long distance, the putative activated gene may be located distal to the integration junction region. Therefore, we decided to clone the distal regions of the flanking sites by chromosomal walking. We failed to obtain positive clones from the Lambda T5 library, when we rescreened this library with the CSP2 probe. A NIH3T3 library was used instead since NIH Swiss mice are closely related genetically to Balb/c mice. Two different clones were isolated. Clone 11 consisted of an 18.2 kb genomic fragment and clone 32 consisted of a 14.5 kb genomic fragment. Restriction mapping analyses revealed that the genomic fragments of these two overlapping clones covered a 20.5 kb region at the bcnc-1 locus, with about 10.5 kb flanking sequence at each side of the viral integration (Figure 2). Three SacI fragments derived from clone 11 and clone 32 were subsequently cloned into plasmid vectors to perform finer restriction mapping. The results obtained from restriction mapping these subclones and the original clones correlated well, and yielded the map illustrated in Figure 2. Northern analysis was repeated with probes to the unique regions within the 20.5 kb region (Figure 2E). RNAs from different tissues were included for the detection of transcripts. Still, no mRNA was detected. A :91: ‘ I if E MoMuLV env 1 a a e i“ t 'i 11‘ iii if“ c 1 1 I x a ' “ p e C L l i ‘3 D 'I 'I can ‘4 1kb E cam-i 1‘11 iii? ITITIII iiiI...rm 1““ E:- ‘uh fl bent-l s— — “algae Figure 2. Cloning strategy and restriction endonuclease mapping of the bone-1 locus. (A) Map of MoMuLV. (8) Map of the clone C isolated from the lambda T5 library. (C) Map of the CS fragment This env-containing Bglll DNA fragment was subcloned for the isolation of the flanking cellular region. (D) The Pstl fragment (03P2) containing the flanking cellular region was subcloned for use as a hybridization probe. (E) Map of genomic DNA from the bone-1 locus. The big arrow represents the site of viral integration in Tumor 5. Restriction fragments containing sequences unique to this region are indicated by filled boxes. Restriction endonucleases: B, BamHl; E, EcoRl; G, Bglll; H, Hindlll; K, Kpnl; N, Notl; P, Pstl; Sau, Sau3A; S, SacI; X, Xbal. ' 80 Bone-1 sequence was conserved among some species A "200" blot containing DNAs from chicken, rat, human, hamster, bovine, and goat was used to examine the sequence conservation of the bonc-1 locus. Surprisingly, we could detect discrete bands by hybridization with C3P2 in all of the species after washing to a stringency of 0.2 x SSPE at 65° C (Figure 3) despite the fact that no mRNA was detected in this region. Sequence analyses of bone-1 The sequence conservation suggested that bone-1 might contain important gene sequences. We therefore sequenced the C3P2 fragment. Figure 4 shows the DNA sequence of the entire 1327 bp 03P2 fragment, with some uncertainty in nucleotide numbers 444 and 446. This uncertainty is due to the strong pause of the sequence reaction at this region. Several methods including the use of ITP and temperature modification in the sequencing reactions have been used to resolve this sequence ambiguity without success. Sequences of the CSP2 fragment were compared to those in the GeneBank and EMBL databases. No homology was found (data not shown). When the sequence was examined for the presence of possible open reading frames (ORFs), several possible ones were identified (Figure 5). The longest ORF, from nucleotide 565 to 957, encoded 131 amino acids. When the protein sequences of these putative ORFs were examined for the presence of known functional domains (motifs) such as leucine-zipper, no known protein motifs were found (data not shown). 81 23> i, j: 9.4» ’ . ;. 66>: f 3 4,. I”, are U . 5“? 5' W: ,3) , 4.4»: ‘ ‘1 '. a 2-3'3 . Figure 3. bonc-1 is evolutionary conserved. Southern blot analysis of DNAs from human (lane 1), hamster (lane 2), bovine (lane 3), goat (lane 4), chicken (lane 5), and rat (lane 6). DNA was digested with EcoRI and 10 pg of each sample was electrophoresed through 0.8 % agarose. The blot was probed for bone-1. Size markers are the positions of an ethidium bromide-stained Hindlll digest of bacteriophage l, and are denoted in kilobases. 82 GCCCTCGCCGGGACGACGGGCGCCTCCGGAGCCGGGGCCGCGGCCGAGGCGCCGA CGCCGCGCGAGCTCGCCCAGACGGGGGCCCGCCCAGCCTGTCAGCGCGGCGGGAG GCCGGCGGTGGCCGCTGCTGCCCCCTTTCTGCTCTGCCTCAGCTTCCTGCAGCCC ACGCCACGCCCACCGCGCGCCGAGCCCGTCTCCGCCCTCCAGGGGCCGCACGCCG CCTCCGCTCGGCCCCGCGACGCCGGCTCAGCTGCCCTGCTCGGCGGGCTCCGGCG CGGTGCAGCTTCGGGAAAGCGGCCCCGAGCGGCGGACGGGGCGTGGGCAAGCCGG CGGGGGTTGGCGGGGGGGCGGAGGCGGCACGCCCCACTGCGCCTGCGCGCCCGCG GCCGCTCTGCGGGCTGGGCCGGGACGGGGAGGCGGCCGCGGGCTCCGGGGAAGCG GAGNCNCGCGTGGAGATTCCCGGGGCAGCCCCCGCGAGAGCGCGCGAGGAGGAGG AGGAGCGGGGCGGGTGCGGGTTGGCGCAGCGTGCTTGCGGCCTCGGCTCGCGGAG GAGACGGCTGGGAATGAGTCAGCCCGGCCGGGAAGGCCCCGCTGCGTCCGAGCTG ATAGGATTGGCGGCGCTGCCTCCGCGGAGACTTTCCCCTTCCTGCTTTCGTTTCC ACAGGCGGCTCCCTGCCCTAAGCGCTCGGCCCAGGGGACGTGGCACCGTGGACCG GGCGCTGAGACCCAAGTACCTGACTTCAGTTAACCACCACTTCTGCGAAGGGACC GCTTGGAAGAAGAAACGACGTATAGGGTCCCAAAGAGCCACGTTCATTCATTCAA GAATTTGTTCCCTGCTAGGCAAGGGCTCCCAGGAGAGGAGACGAAGGCAGCACCC ATTCTGCATGTGCTATTCTCATAGAAGCCAGAAAAGAGGCTTTAAATCTGCACTG CTCTTTGTGTGTGATTGCCAGTAACGGGTACACACACACACACAAAGGTGAGTGG AGATGTTATTTTGAGATGACCAGAAAAGACCTCTTAGAGTGGCATTTGAGCAGAA ATGTGAAGGATGAGAACGATCCAGTCCTGGAACTATCTGGTCAAGAAAGAAGCTA ACAATGAAAAGGATTCTGAAAAGGGAATGTGTTTTAAGGACCAAGAAAGAACCCT CAATGGAAGACAGTAAAAGGAAAGGGGCTAAATAAGAAGACAAAGAAATAACTGG GGTCATTCTGTATAGGGCTTCTTATTTTTTCTCTTAGCCCCAGGTTTGTCAACCG AAATGAAGCATTCACAACACCCACGTCCCCTTATATACGTGACGGATGCTGTTAT TCAGATC 55 110 165 220 285 330 395 440 495 550 605 660 715 770 825 880 935 990 1045 1100 1155 1210 1265 1320 1327 Figure 4. DNA sequences of the C3P2 fragment. X indicates undetermined nucleotides. EKB soan.ao. u. on 05.3 3 Co a 8.9: 9.53.. coco 2:. «notes: «98.. E .0» .5 pea-050.3 9.2. 38...... 332.3 u use... 2.63.. coco of 432033.. admit—mu: 32:3,. Eon 8330303.. 053s mo— mo 403633: 53.33 «2630.. Bus 3...... 3. to 329.3 < 8.6.; 3:80.. coco of 2.2! mean... 9.53.. coco 309.3 09.5 of e... u use. a .< .333 539.253 E3959. .89 p23 of .2863 533:5 9.30.59. xon 3.3 of gone 9.3 .3223 of .23.: 3:63.. code so c233 30:02.0... 2: 695.... 9.33.. 3338 5a of acomocooc n- oca .~- .7 .n .N J .sstooad c3303”. 9:30.. coco £5.59; ~anu of co seconds... as. E «95.... 9.33.. coco 0383:... .m 1 _ _ _ __ _ _:-:.o._ '..-...o...._ _ _ ml EBB...— __ —. =— ”U _ _ NI _ . _ _ _ 1 _ 1.2:...— _ _ MI 3— _ _ F3438...— _ _d_ _ _ ‘_ m .3. _ :................_,___ _ _ _ _ N _ _ _ __ _ _ . . — — —P-Ie=e.eeefle— 03.33 339.3 .ucao. :33 one...» o..u.uc. ~ .eodecoo e5 otsm_m Frede fragrr abou1 abou1 There (ngr anec fieque that g maDD to Chr D iympr RFLP restric {Nobe ShOWr chm Fl9Ure 84 Bone-1 was mapped to chromosome 19 by RFLP analyses In collaboration with Marge Strobel, Neal Copeland and Nancy Jenkins (NCl, Frederick), the bonc-1 locus was mapped to mOuse chromosome 19 by restriction fragment length polymorphism (RFLP) analyses. Bone-1 was mapped to a region about ,-_16.0 t 2.7 cM from Porno-2 and 2.3 i 1.6 cM from cypzc. Pomc-2 is H ._ about, 6.0 1- 1.7 cM from Ly-1, and CypZC is about 2.3 1.3 cM from Tdt. Therefore, bonc-1 was 4.7 i 3.0 cM from Tdt, and 22.0 i 4.5 cM from Ly-1 (Figure 6). Two other loci were also mapped near this region: Ipr, a genetic loCus linked to a recessive lymphoproliferatiOn'trait (Lyon and Scarle, .1989), and a frequent virus integration site, gin-1 (Villemur et al., 1987). RFLP analyses showed that gin-1 is not identical to bone-1 (Figure 6). Watanabe et al. (1990) have- mapped Ipr to 6.1 cM from Tdt and 19.3 cM from Ly—44, a locus physically mapped to chromosome 11q12-13 which is very close to Ly-t in chromosome 11513. Due to the close linkage of bone-1 to Ipr and their common association with . lymphoid disorders, we examined the possibility of both loci being identical by an RFLP analysis. DNA of Balb/c.lpr mice and Balb/c were digested with 7 different restriction enzymes, and subjected to a southern hybridization with the C3P2 probe. No DNA rearrangements were found in DNA of Balb/c.lpr mice (data not shown). ll-le-u 5 cM [gnu-2 lens-l meta-I Ii! Chromosome 19 Figure 6. RFLP mapping ofbonc-1. )i J! (U 85 Discussion: A tumor-specific viral integration site (bone-1) which may be associated with the growth advantage of tumors derived from the R2 cell line was isolated. Sequence homology to other species was found within this locus, significance of which remains unknown. Similar results have been observed at other loci: evi-1, which is a frequent viral integration site found in AKXD murine myeloid tumors (Mucenski et al., 1988a); and dsi-t, which is also a frequent viral integration site identified from Fisher rat thymomas (Vijaya et al., 1987). Evl-1 was later found to encode a zinc finger protein (Morishita et al., 1988), whereas no transcripts have been associated with dsi-1 thus far. Although no mRNA was found over a region extending 10 kb to each side of the site of viral integration, we cannot rule out the possibility of the activation of a gene beyond this 20 kb region. Long distance activation of the myc protooncogenes has been reported by Lazo et al. (1990). They found that provirus integrations 30 kb 3’ of c-myc (mlvi-4) and 270 kb 3’ of c-myc (mlvi-1) enhanced c-myc expression in rat T lymphomas. Further chromosomal walking may resolve this question. it is also possible that the tumor-specific viral integration at this locus is coincidental with another true activation event. For example, there are still three or four tumor-specific viral integration sites remaining to be analyzed. Nevertheless, the conservation of the bone-1 locus across species and the presence of open reading frames leaves open the possibility that it may represent a gene expressed at low levels, restricted to a tissue type, or restricted temporally. Bone-1 was mapped to mouse chromosome 19 near a previously described 86 frequent viral integration site, gin-1 (Villemur et al., 1987), and the Ipr locus which is linked to a recessive lymphoproliferation trait (Lyon and Scarle, 1989). Results from RFLP analyses suggest that bone-1 and gin-1 are not identical. Our results from RFLP analyses on the DNA of Balb/c and Balb/c.lpr mice did not suggest or exclude the possibility of bone-1 and Ipr being identical. However, because Ipr is linked to a recessive trait and we only detected the gene rearrangement of bone-1 on one allele, they are likely to be non-identical. The fact that it is close to the Ipr locus provides a use for this locus in the search for genes responsible for the Ipr lymphoproliferation trait. RE‘ 86'. mi: IUTT Chl Hit the Ihl 87 References: Aaronson S. (1991). Growth factors and cancer. Science 254, 1146-1153. Auffray C. , and F. Rougeon. (1980). Purification of mouse immunoglobulin heavy—chain messenger RNAs from total myeloma tumor RNA. Eur. J. Biochem. 107, 304-314. Aviv l-l., and P. Leder. (1975). Purification of biologically active globulln message RNA by chromatography on oligothymidylic acid-cellulose. Proc. Natl. Acad. Sci USA 69, 1408. Baumbach W.R., E.M. Colston, and MD. Cole. (1988). Integration of the Balb/c ecotropic provirus into the colony-stimulating factor-1 growth factor locus in a myc retrovirus induced murlne monocyte tumor. J. Virol. 62, 3151-3155. Chen S.-C., D. Redenius, and RC. Schwartz. (1991 ). Tumorigenesis of a v-Ha-ras-expressing pre-B cell line selects for c-myc activation. Biochem. Biophy. Res. Comm. 178, 1343-1350. Cuypers H.T., G. Selten, W. Quint, M. Zijlstra, E.R. Maandag, W. Boelens, P. van Wezenbeek, C. Melief, and A Bems. (1984). Murine leukemia virus-induced T cell lymphomagenesis: integration of proviruses In a distinct chromosomal region. Cell 37, 141-150. Devereux J., P. Haeberl, and O. Smithies. (1986). A comprehensive set of sequence analysis programs for the VAX. Nucl. Acids Res. 12, 387-395. Dickson C., R. Smith, S. Brookers, and G. Peters. (1984). Tumorigenesis by mouse mammary tumor virus: proviral activation of a cellular gene in the common integration region int-2. Cell 37, 529-536. Fung Y.-K.T., W.L Louis, LB. Crittenden. and H.J. Kung. (1983). Activation of the cellular oncogene c-erbB by LTR insertion: Molecular basis of erythroblastosls by Avian Leukosis Virus. Cell 33, 357-368. Hayward W., E.G. Neel, and S. Astrin. (1981). Activation of a cellular onc gene by promoter insertion in ALV-induced lymphoid leukosis. Nature 290, 475-480. Hicks G.G.. and M. Mowat. (1988). integration of Friend Murine Leukemia Virus into both alleles of the p53 oncogene in an erythroleukemia cell line. J. Virol. 62, 4762-4755. lhle J.N., 8.8. White, B. Sisson, D. Parker, D.G. Blair, A Schultz, C. Kozak. R.D. Cunsford. D. Askew. Y. Weinstein, and R.J. lsofort. (1989). Activation of c-Ha-ras protooncogenes by retrovirus insertion and chromosomal rearrangement in a Moloney leukemia virus-induced T cell leukemia. J. Virol. 63, 2959-2966. Lazo P.A. , J.S. Lee, and RN. Tschilis. (1990). Long-distance activation of the myc protooncogene by provirus insertion in MIvi-1 or MIvi-4 in rat T-cell lymphoma. Proc. Natl. Acad. Sci. USA 87, 170-173. Lyon M.F., A.G. Scarle. (1989). Genetic variants and strains of the laboratory mouse. 2nd ed. pp. 209. Morishita K., 03. Parker, M. Mucenski, N.A. Jenkins, N.G. Copeland; and J.N. lhle. (1988). Retroviral activation of a novel gene encoding a zinc finger protein in lL-3 dependent leukemia cell lines. Cell 59. 831 840. Mucenski l (1988a) ldi myeloid tut Muller W J product ac Nusse R. - contain a t Payne GS Sambrook Spring Har Sanger F ., Proc. Natl Schwartz l lambda ch Schwartz i am V-Ha-l 3221-3231 Selten G.. and A Be homology locus hyt 1077-1 030 Siocking ( genes inv Williams ( TSUkamot‘ "‘9 inll 309!)er Win 8. 'nsemons com"ion DNA Moi.F Chm-”080' 88 Mucenski M.L, B.A. Taylor, J.M. lhle, J.N. Hartley, H.C. Morse, Ill, N.A. Jenkins, and MG. Copeland. (1988a). Identification of a common ecotropic viral integration site evi-1 in the DNA of AKXD murlne myeloid tumors. Mol. Cell. Biol. 8, 301—308. Muller W.J., F.S. Lee, C. Dickson, G. Peters, P. Pattengae, and P. Leder. (1990). The int-2 gene product acts as an epithelial growth factor in transgenic mice. EMBO J. 9. 907-913. Nusse R., and HE. Varmus. (1982). Many tumors induced by the mouse mammary tumor virus contain a provirus integrated in the same region of the host genome. Cell 31, 99-109. Payne G.S., J.M. Adams, and HE. Varmus. (1982). Multiple arrangements of viral DNA and an activated host oncogene in bursal lymphomas. Nature 295, 209-213. Sambrook J., E.P. Fritsch, and T. Manlatls. (1989). Molecular cloning: A laboratory manual, Cold Spring Harbor Laboratory . (New York: Cold Spring Harbor). Sanger F., S. Nicklen, and AR. Coulson. (1977). DNA sequencing with chain-terminating inhibitors. Proc. Natl. Acad. Sci. USA 74, 5463-5467. Schwartz R.C., G.E. Sonenshein, A. Bothweli, and ML. Gefter. (1981). Multiple expression of lg lambda chain encoding RNA species in murine plasmacytoma cells. J. immunol. 126, 2104-2108. Schwartz R.C., LW. Stanton, S.C. Riley, K.B. Marcu, and ON. Witte. (1986a). Synergism of v-myc and v-Ha-ras in the in vitro neoplastic progression of murlne lymphoid cells. Mol. Cell. Biol. 6, 3221-3231. Selten 6., HT. Cuypers, W. Boelens, E. Robanus-Maandag, J. Verbeek, J. Domen, C. van Beveren and A Bems. (1986). The primary structure of the putative oncogene pim-1 shows extensive homology with protein kinase. Cell 46, 603-611. ShenCng G.LC., M. Potter, J.F. Mushinski, S. Lavu, and E.P. Reddy. (1984). Activation of the myc locus hybridization insertional mutagenesis in piasmacytoid iymphosarcomas. Science 226, 1077-1080. Stocking C., C. L6llger, M. Kawai, S. Suciu, N. Gough, and W. Ostertag. (1988). Identification for genes involved in growth autonomy of hematopoietic cells by analysis of factor-independent mutants. Cell 53. 869-879. Tsukamoto A.S., R. Grosschedl, R.C. Guzman, T. Parslow. and HE. Varmus. (1988). Expression of the inf-1 gene in transgenic mice is associated with mammary gland hyperplasia and adenocarcinomas in male and female mice. Cell 55, 619-625. Vijaya S., D.L. Steffen, C. Kozak, and H.L Robinson. (1987). A region with frequent proviral insertions in Moloney murlne leukemia virus induced rat thymomas. J. Virol. 61, 1164-1170. Villemur R., Y. Monczak, E. Rassart, C. Kozak, and P. Jolicoeur. (1987). identification of a new common proviral integration site in cross passage a murlne leukemia virus-induced mouse thymoma DNA. Mol. Cell. Biol. 7, 512-522. Watanabe T., A. Shimizu, and Y. Sakai. (1990). A molecular genetic lineage map of mouse chromosome 18 and 19. Fourth international workshop on mouse genome mapping. Abstrct 101. Chapter 4 IL-7 expression in a v-Ha-ras transformed pre-B cell line is not sufficient for tumorigenicity: differing assessments in clonal versus heterogeneous populations Shu-Chih Chen,1 Diane Redenius,1 Judy C. Young,2 and Richard C. Schwartz‘ ‘Department of Microbiology, Michigan State University, East Lansing, MI 48824-1101 2Department of Microbiology and Molecular Genetics, University of California-Los Angeles, Los Angeles, CA 90024-1570 89 Abstract: A recombinant retrovirus expressing IL-7 was superinfected into a murine pre-B cell line previously infected with a retrovirus expressing v-Ha-ras. Populations of cells polyclonal for integration of the iL-7 virus and independent of iL-7 for growth were generated. lL-7 expression caused only a minimal enhancement of tumorigenicity. Surprisingly, the introduction of v—myc expression also had only a minimal effect. in contrast, co-infection of murine bone marrow with retroviruses expressing iL-7 and v-Ha-ras generated clonal B lymphoid outgrowths that were tumorigenic. We propose that although iL-7 and v-Ha-ras may contribute to tumor progression, their introduction into a heterogeneous population of bone marrow cells allows the selection of additional oncogenic events occurring independently in that population. The oncogenic potential of lL-7 expression, in itself, and deregulated expression of other genes is probably best assessed in well-characterized individual cell lines. 91 Introduction: The neoplastic transformation of hematopoietic cells is generally accepted to be a multi-step process (Schwartz and Witte, 1988). This process can involve the overexpression of hematopoietic growth factors, as well as events involving other classes of oncogenes (Sawyers et al., 1991). For example, a translocation activating lL-3 expression has been observed in a form of human acute pre-B leukemia (Meeker et al., 1990). Overexpression of lL-3 by itself has been shown to cause a myeloproliferative disorder rather than leukemia (Chang et al., 1989; Wong et al., 1989) suggesting that its role in leukemogenesis requires cooperation with other events. iL-7, a cytokine for pre-B cells (Namen et al., 1988) and T cells (Morrissey et al., 1989), has recently been examined for its role in the transformation of pre-B cells. Young et al. (1991) found that hyperexpression of lL-7 in a pre-B cell line was neither necessary nor sufficient for neoplastic transformation. Although the cell line’s dependence upon exogenous iL-7 was alleviated, its growth in soft-agar medium was not enhanced and only one of six IL-7-expressing derivatives of the cell line was tumorigenic. In contrast, Overell et al. (1991) found that a pre-B cell line made growth factor-independent by hyperexpression of lL-7 was tumorigenic. However, their data indicated that events in addition to lL-7 expression were required for factor independence. These seemingly contradictory findings leave unresolved the potential for autocrine lL-7 expression to contribute to the tumor progression of pre-B cells. On the other hand, both studies suggest that hyperexpression of lL-7 may have a role in the cooperative transformation of pre-B cells with other oncogenes. in previous studies, we found that v-Ha-ras-expressing pre-B lymphoid cells 92 displayed an intermediate transformed phenotype and were infrequeme tumorigenic (Schwartz et al., 1986 a, b). The tumors that did arise were not dependent upon IL-7 for growth, unlike the pre-B cells from which they arose (unpublished observations, Chen and Schwartz). This led us to investigate whether high level autocrine expression of lL-7 would be sufficient for the tumorigenicity of a v-Ha-ras-expressing pre-B cell line. Since we had previously demonstrated the cooperativity of v-myc and v-Ha-ras in pre-B cell transformation (Schwartz et al., 1986 a, b) and a role for myc activation in the tumor progression of a v-Ha-ras-expressing pre-B cell line (Chen et al., 1991), we directly compared the oncogenic potential of lL-7 and v-myc in a v—Ha—ras-expressing pro-B cell line. While expression of lL-7 may partially alleviate dependence on exogenous growth factors for growth, it is clearly not sufficient for tumorigenesis. Curiously, we have found a disparity for both lL-7 and v-myc between the consequences of introducing their expression into a v-Ha-ras-expressing cell line and the simultaneous introduction of these oncogenes by co-infection of primary bone marrow cells. Pre-B cell lines derived by co-infection of bone marrow were fully tumorigenic (see results reported here and Schwartz et al., 1986a), while lines derived by sequential addition to a cell line were not. Neither lL-7 nor v-myc in combination with v-Ha-ras are truly sufficient in and of themselves for cooperative transformation of pre-B cells. 93 Results: Superlnfection of a v-Ha-ras-expressing pre-B cell line in order to test the oncogenic potential of IL-7 expression in cooperation with v-Ha-ras expression, we generated a v-Ha-ras-expressing cell line that could easily be superinfected by a retrovirus carrying the gene for IL-7. Fresh murine bone marrow was infected with a helper-free stock of SV(X)-Ha-ras, a v-Ha-ras- expressing retrovirus (Schwartz et al., 1986a), and the bone marrow was placed under long-term B cell culture conditions (Whitlock and Witte, 1982). A clonal pre-B cell outgrth designated w2R4 resulted from this infection. This cell line expresses v-Ha—ras from a single proviral integration site and was found to be dependent on a cellular feeder layer or IL-7 for growth, unable to grow in soft agar medium, and not to be tumorigenic (data not shown). ¢2R4 was subjected to three different retroviral infections: 1. An lL-7-expressing retrovirus, IL-7(SH) (Young et al., 1991) and Moloney murine leukemia virus (MoMuLV). 2. A v-myc-expressing retrovirus, MMCV-neo (Wagner et al., 1985) and MoMuLV. 3. MoMuLV. Three weeks post infection, DNA and RNA were isolated from the infected cultures and analyzed to assess infection and integration of the above mentioned viruses and to verify expression of v-Ha-ras, iL-7 and v-myc in the infected cells. Southern blots of DNAs isolated from two cultures each of MoMuLV, “IL-7" and “v-myc" infected cells were hybridized with probes for iL-7 (Figure 1A and B), v-myc (Figure 1C and D) and v-Ha-ras (Figure 1E). Hindlil digestion (Figure 1A) Figure 1. Proviral integration. DNA (10 pg) was digested with the indicated restriction enzymes, electrophoresed through 0.8% agarose, and transferred to nylon membranes. Lane 1, NiH3T3/lL-7(SH); lane 2, $2R4/MoMuLV-1; lane 3, w2R4/MoMuLV-2; lane 4, w2R4/Myc-1; lane 5, w2R4/Myc-2; lane 6, t2R4/lL7—1; lane 7, w2R4/lL7—2; lane 8, RM2. Size markers are the positions of an ethidium bromide-stained Hindlli digest of bacteriophage lambda DNA. (A) HindIII-digested DNA was hybridized with an IL—7 probe. The position of the retroviral iL-7 gene is marked at 0.47 kb. (8) BamHI-digested DNA was hybridized with an IL-7 probe. (C) BamHi-digested DNA was hybridized with a v—myc probe. The position of the retroviral v—myc gene is marked at 2.5 kb. (D) Hindlii-digested DNA was hybridized with a v-myc probe. (E) EcoRl-digested DNA was hybridized with a v-Ha-ras probe. The position of the v—Ha—ras integration-related restriction fragment is marked at 7 kb. 95 Figure 1. Proviral integrations. A: IL'7/Hll‘ld III C: MYC/Baml-l I 1 2 3 4 5 s 7 a 23»: I ‘5 " “ i 9.4} 6» -u'L-.'.g,-.i D: MYC/Hlnd III 2 3 4 5 (I ‘1' . 678 96 revealed the diagnostic viral 0.47 kb lL-7-specific restriction fragment in both iL-7-infected cultures (lanes 6 and 7), as well as in the NIH3T3 cell line that produced the iL-7 virus (lane 1). The intensity of hybridization to this fragment is as great in the infected cultures as it is in the NiH3T3 virus-producing cell line, suggesting a high proportion of infected cells. The other restriction fragments represent the endogenous lL-7 gene. BamHl digestion, which cleaves once within the IL-7 virus genome, was used to assess the number of viral integrations and the clonaiity of the iL—7-infected populations. This analysis (Figure 1B) revealed the lL-7-infected cultures to contain multiple viral integration-related restriction fragments of less than haploid abundance (lanes 6 and 7) indicating polyclonal populations of IL-7-infected w2R4 pre-B cells. All cells possessed two iL-7-specific restriction fragments of “'4 kb and “18 kb. Rehybridization of the BamHl-digested DNAs with a v-myc probe (Figure 10) revealed a 2.5 kb restriction fragment diagnostic of v-myc infection in those infected populations (lanes 4 and 5), as well as in RM2 (lane 8), a v-Ha-ras/v-myc transforrnant previously characterized (Schwartz et al., 1986a). The restriction fragment of ~6 kb represents endogenous c-myc. The v-myc infected populations were evaluated for viral integration by rehybridization of the HindiIl-digested DNAs with a probe for v-myc (Figure 1D). Hindlli cleaves the v-myc viral genome only once. Similarly to the case of iL-7 infection, the v-myc-infected cultures (lanes 4 and 5) were shown to be polyclonal with respect to v-myc viral integration with a smear of v-myc-specific restriction fragments as opposed to the single viral fragment in the clonal RM2 cell line (lane 8). The "5 kb restriction fragment represents endogenous c-myc. in order to verify that the outgrowths in the infected cultures were all derived from ¢2R4, the 97 v-Ha-ras viral integration sites were compared among the cultures. An EcoRl digestion cleaves once within the viral genome to yield integration site specific fragments. All of the infected cultures contained the same size v-Ha-ras-related restriction fragment of ~7 kb confirming their derivation from ¢2R4 (Figure 1E, lanes 2-7). The ”20 kb restriction fragment represents the endogenous c-Ha-ras gene. Cytoplasmic RNAs of the infected ¢2R4 cells were examined by Northern hybridization analysis. The expected genome-length retroviral transcript of "5.4 kb was observed in all the populations with a probe for v-Ha-ras (Figure 2A). Rehybridization with an IL-7 probe revealed the expected 4.0 kb genome-length retroviral transcript in both iL-7-infected populations (Figure 2B, lanes 5 and 6) and a v—myc probe showed the expected genomic "8.0 kb and subgenomic "5.5 kb RNAs in both v-myc-infected populations (Figure 2C, lanes 3 and 4). Rehybridization to a probe for rat giyceraldehyde phosphate dehydrogenase (GAPDH) verified that similar quantities of RNA were analyzed among the various infected populations (Figure 2D). Therefore the levels of mRNA expression for v-Ha-ras, v-myc and lL-7 were roughly equivalent between populations infected with viruses expressing those genes. The superinfected o2R4 populations consist of pro-B cells Given that the activity of lL-7 within the B lineage is restricted to pre-B cells, it was important to confirm that the superinfected cells had maintained the pre-B phenotype of ¢2R4. Southern hybridization analysis of EcoRI-digested DNAs revealed that all of the populations were identically rearranged in their mu heavy FeruFLICleraHdli. 1 2 3 4 5 6 A: RAS “fiuflnb 42885' e: lL-7 i l (x IWYC E. El ‘__55 "A 9 ‘ ; 428$ ‘w‘w-Hi urn-V4 mwmmvv wt} 'Huflufl HALAszJ EL: 31""V‘h‘ «185 o: GAPDH .__15 Figure 2. Retroviral transcription. Cytoplasmic RNA (20 pg) was denatured, electrophoresed in a 1% agarose-formaldehyde gel, and transferred to a nylon membrane. Lane 1, w2R4/MoMuLV-1; lane 2, ¢2R4/MoMuLV-2; lane 3, 1|:2R4/Myc-1; lane 4, 1|:2R4/Myc-2; lane 5, w2R4/lL7-1; lane 6, w2R4/IL-2. The positions of ethidium bromide-stained 28S and 183 rRNAs are marked. (A) Hybridization with a v-Ha-ras probe. The position of the SV(X)-Ha-ras genome-length RNA is marked at 5.4 kb. (B) Hybridization with an lL—7 probe. The position of the IL-7(SH) genome-length RNA is marked at 4.0 kb. (C) Hybridization with a v—myc probe. The positions of the MMCV-neo genome-length and subgenomic v-myc RNAs are marked at 8.0 and 5.5 kb, respectively. (D) Hybridization with a GAPDH probe. The position of GAPDH mRNA is marked at 1.5 kb. Sti en Wt wit! Con 99 chain locus (Figure 3A), again verifying that all of the cells were derived from 1p 2R4. Analysis of BamHI-digested DNAs revealed all of the populations to have agermline configuration in the kappa light chain locus (Figure 38) confirming a pre-B phenotype. This confirmed that the cells should indeed retain responsiveness to lL-7. lL-7 and v-myc infected populations show enhanced growth factor independence Transformation in a complex culture system, such as that employed here where there are both nonadherent lymphoid cells and adherent stromal cells, requires one to distinguish between effects mediated by events occurring in the lymphoid cells versus those occurring in the stromal layer. To that end, the infected populations were examined for growth in liquid culture without an adherent stromal layer (T able 1). While the MoMuLV-infected controls showed no growth, or rather indolent growth, at plating densities up to 5x104 cells/ml, the IL-7- and the v-myc-infected populations displayed a 3 to 8 fold increase in growth over the MoMuLV infected control when seeded at 5 x 10‘ cells/ml. At 10’5 cells/ml, the differences in growth were not nearly as apparent suggesting the autocrine stimulation of growth factors other than lL—7. lL-7-infected cells do not show enhanced growth in soft agar medium, while v—myc-infected cells do (T able 2). When plated in soft agar medium over a cellular feeder layer, populations infected with iL-7 had a plating efficiency of about 0.1% (Table 2). This is actually less than the plating efficiency of the MoMuLV-infected control populations, about 0.2%. In contrast, v-myc infection elevated the plating efficiency of it: 2R4 about 5-foid to 1%. A:.iH 1234567 23»- 94> 6.6» . 4.4>' -’. . . -' 1 n ' . . . . -. 1'- .. $3- u-.. 1-3.. . «M Figure 3. Immune loci define a pre-B phenotype. DNA (10 ng) was digested with the indicated restriction enzymes, electrophoresed through 0.8% agarose, and transferred to nylon membranes. Lane 1, fibroblasts; lane 2, i|r2R4/MoMuLV-1; lane 3, w2R4/MOMuLV-2; lane 4, 1|r2R4/Myc-1; lane 5, w2R4/Myc-2; lane 6, w2R4/lL7—1; lane 7, ¢2R4/|L7-2. Size markers are the positions of an ethidium bromide-stained Hindlll digest of bacteriophage lambda DNA. (A) EcoRl-digested DNA was hybridized with a probe for heavy chain J regions. (B) BamHl-digested DNA was hybridized with a probe for the kappa light chain constant region. Table 1. Cell population RM2 t 2R4/MoMuLV-1 it 2R4/MoMuLV-2 t 2R4/Myc-1 1r 264/Myc-2 t 2R4/IL7-1 t 2R4/IL7-2 RIL7.1 RIL7.2 101 Growth without cellular feeder layers. initial Density13 5 x 103/ml 1 x 104/ml 5 x 104/mi 1 x105/ml Density on Day 7 2.2 t 0.1(106) 4.0 a: 0.2(106) 6.6 i 0.500“) 7.4 i 1.000") < 103 < 103 5.3 : 0400‘) 2.0 t 0000“) < 103 1.4 : 0.0(103) 6.0 : 0.5(105) 2.1 : 0.300“) 1.0 : 0.0(105) 4.5 i 0500‘) 2.0 : 0.000“) 3.6 i 0.3(106) 2.6 : 2.6(103) 2.1 : 0.7(103) 1.7 .1: 0.2(105) 3.6 x 0000“) < 103 7.7 :t 35003) 2.3 1' 0.300“) 4.2 1 0.200") 1.0 t 0.8(103) 3.6 .4.- 0.0(103) 4.2 i 0.0(105) 5.7 : 0.5(106) NDb 2.6 : 0.0(103) ND 2.0 : 0.2(105) ND 2.0 : 0.2(105) ND 1.6 : 0.1(105) 3Cells were plated onto 6 cm culture dishes in 4 ml media. On day 4, each dish received 2 ml fresh medium. Values are the average (with the range) of duplicate cultures. bNot done. Table 2. Soft agar growth and tumorigenicity. Cell population % Cloning efficiency Animals with tumors/ Average days until in soft agar animals tested dead or moribund RM2 >5 13/14° 25 t2R4/MoMuLV-1 0.26 0/6b - t2R4/MoMuLV-2 0.20 0/6b -- t2R4/Myc-1 1.06 1 /8b 42 1: 2R4/Myc-2 0.64 1 /6'° 73 t2R4/lL7-1 0.09 3/6" 36 t2R4/IL7-2 0.10 0/6b - RIL7.1 ND“ 3/4" 53 RIL7.2 ND 7/8d 40 “Not done. bThese values have no statistically significant differences among them by Fisher's exact test (p s 0.05). °This value is statistically different from those in b by the same test in b. dThese two values are combined, and the resulting value is statistically different from those in b by the same test in b. 102 A positive control of RM2 cells had a plating efficiency greater than 5%. Curiously, cells produced by the introduction of v-myc expression subsequent to v-Ha-ras expression have poorer plating efficiencies in soft agar than cells produced byco-infection of primary bone marrow cells (is. RM2; Schwartz et al., 19866) The lL-7 and v-myc-infected populations are not tumorigenic The abilities of the superinfected 1|:2R4 populations to form tumors In vivo was tested by intraperitoneal injection of these cells into syngeneic BALB/c mice. While positive control RM2 cells formed tumors quite consistently, both the v-myc- and iL—7-infected populations exhibited no statistically significant difference in tumorigenicity from the MoMuLV-infected 1|:2R4 cells with Fisher’s exact tests (Table 2). These results suggested that neither lL-7 nor v-myc were not capable of cooperating with v-Ha-ras in the induction of B cell tumors. Co-infectlon of primary bone marrow cells with lL-7 and v-Ha-ras retrovirus yields clonal outgrowths that are tumorigenic The surprisingly low tumorigenicity of v-myc-infected populations of a v—Ha—ras-expressing pre-B cell contrasts sharply with our earlier observation of highly tumorigenic cell lines generated by the co-infection of primary bone marrow cells with v-Ha-ras and v-myc retroviruses 0.6. RM2; Schwartz et al., 19866). We therefore decided to examine the effects of co-infecting freshly explanted murine bone marrow cells with v-Ha-ras and IL-7 retroviruses. Bone marrow cells from BALB/c mice were infected singly with the lL-7 retrovirus, the v—Ha—ras retrovirus or doubly infected with both retroviruses. These cells were then plated in liquid 103 culture by the procedure of Whitlock and Witte (1982). In 20 co-infections, only two cell lines containing both lL-7 and v—Ha—ras retroviral integrations were obtained (RIL7.1 and RIL7.2) after about 2 months in culture. Other outgrowths contained only the v-Ha-ras retrovirus. Single infections with the lL-7 retrovirus never resulted in outgrowths containing that virus, while single infections with the v-Ha-ras retrovirus consistently produced v-Ha-ras-expressing cell lines similar to those previously reported (data not shown; Schwartz et al., 19866). Southern blot analysis of DNAs isolated from RIL7.1 and RIL7.2 revealed these outgrowths to be clonal, unlike the isolates derived by superlnfection of1p2R4. BamHl digestion (Figure 4A) revealed one iL-7 viral integration-related restriction fragment for RIL7.1 (lane 1) and two integration-related fragments for RIL7.2 (lane 2). DNA from uninfected (lane 3) and infected cells possessed two endogenous lL-7-specific restriction fragments of “4 kb and ~18 kb. EcoRl digestion of the DNAs (Figure 48) showed one v-Ha-ras-specific viral integration-related restriction fragment for RIL7.1 (lane 1) and two integration-related fragments for RIL7.2 (lane 2) in addition to the endogenous c-Ha-ras fragment of "20 kb. Subcloning of RIL7.2 in soft agar medium demonstrated that the multiple viral restriction fragments represent multiple integrations in a single clone rather than several clones (data not shown). Northern analyses detected v-Ha-ras mRNA in both RIL7.1 and RIL7.2 (Figure 5A, lanes 1 and 2), but detected iL-7 mRNA only in RIL7.2 (Figure 58, lane 2). Southern blot and Northern blot analyses showed RIL7.1 to be a B cell rearranged in both its mu and kappa loci, and expressing the kappa locus (data not shown). RIL7.2 was shown to be a pre-B cell with germline configuration of its kappa DNA (data not shown). Perhaps the progression of RIL7.1 to a B cell phenotype 104 dependent upon factors other than IL-7 for growth eliminated any selective pressure to maintain IL-7 expression in that cell line. RIL7.1 showed only indolent growth when plated without a feeder layer at 105 cells/ml (T able 1). Not surprisingly, RIL7.2, which expresses lL-7 mRNA, was growth factor independent (T able 1). Analysis of the tumorigenicity of RIL7.1 and RIL7.2 revealed these cell lines to be more obviously transformed than populations of ¢2R4 superinfected with the lL-7 retrovirus. Similar to cell lines produced by the co-infection of bone marrow with v-myc and v—Ha—ras retroviruses (i.e. RM2; Schwartz et al., 19866), the two cell lines produced by co-infection with IL-7 and v-Ha-ras retroviruses were highly tumorigenic (T able 2). RIL7.1 produced tumors in three of four animals challenged and RIL7.2 produced tumors in seven of eight animals. 105 A:lL-7 BzRAS _1 2 3 1 2 3 .‘4 Figure 4. Proviral integration. DNA (10 pg) was digested with the indicated restriction enzymes, electrophoresed through 0.8% agarose, and transferred to nylon membranes. Lane 1, RIL7.1; lane 2, RIL7.2; lane 3, uninfected cells. Size markers are the positions of an ethidium bromide-stained Hindill digest of bacteriophage lambda DNA. (A) BamHl-digested DNA was hybridized with an IL-7 probe. (B) EcoRl-digested DNA was hybridized with a v-Ha-ras probe. 106 Figure 5. Retroviral transcription. Cytoplasmic RNA (20 pg) was denatured, electrophoresed in a 1% agarose-formaldehyde gel, and transferred to a nylon membrane. Lane 1, RIL7.2; lane 2, RIL7.1; lane 3, NIH3T3/iL-7(SH); lane 4, NIH3T3/SV(X)-Ha-ras. (A) Hybridization with a v-Ha-ras probe. (B) Hybridization with an lL-7 probe. (C) Hybridization with a GAPDH probe. 107 Discussion: The experiments presented here find autocrine expression of iL-7 insufficient to cause tumorigenicity of a v-Ha-ras-expressing pre-B cell line. This contrasts with previous studies by Overell et al. (1991) that found a pre-B cell line made growth factor-independent by hyperexpression of IL-7 to be tumorigenic. Our results are more consistent with those of Young et al. (1991) that found a pre-B cell line infected with an lL-7-expressing retrovirus to be converted to tumorigenicity only rarely. While it may capable of enhancing growth factor independence of a v-Ha-ras transformant, lL-7 expression had no direct correspondence to tumorigenicity. The rare tumors recovered had a long latency suggesting that ultimately other events were required for tumorigenicity. The lack of lL-7 dependence in tumors derived from v-Ha-ras-transformed pre-B cells is likely to be a consequence of tumor progression rather than its cause. Kremer et al. (1991) have recently found c-src and c-ras to be downstream effectors of signal transduction by nerve growth factor and fibroblast growth factor. if the effects of both lL-7 and v-Ha-ras expression were upon the same signal transduction pathway, cooperativity in transformation might not be expected. Perhaps was is a downstream effector of lL-7, as well as nerve growth factor and fibroblast growth factor. Overell et al. (1991) found the induction of IL-7 independence in a pre-B cell line to be tumorigenic. However, very few of the cells infected with their lL-7-expressing retrovirus actually attained lL-7 independence. Thus, it seems that in their system, too, lL-7 expression is not sufficient for tumorigenicity. Other 108 events are required. Overell et al. (1991) do find that IL-7 independence (as opposed to lL-7 expression) does correlate with tumorigenicity and this result is more difficult to reconcile with our findings. This difference probably lies in the intrinsic properties of the individual cell lines used in the several studies. The v-Ha-ras-transformant used in our studies may require growth factors in addition to iL-7 for optimal growth. A similar v-Ha-ras-expressing pre-B cell line, as well as the cell line used by Young et al. (1991), requires a non-lL-7 factor(s) released by bone marrow stromal cells for Optimal growth (Mulrhead et al., 1990). The cell line used by Overell et al. (1991) may have more simple growth factor requirements, having been selected for its utility in lL-7 assays. Comparing lL-7 directly to v-myc, we found that neither was able to confer tumorigenicity to ¢2R4, although v-myc expression enhanced the ability to grow in soft agar medium. This result was surprising in light of our earlier studies (Schwartz et al., 19866; 1986b), where the in vitro co-infection of bone marrow with retroviruses expressing v-myc and v-Ha-ras yielded fully tumorigenic cell lines. When we performed co-infections of bone marrow cells with lL-7 and v-Ha-ras- expressing retroviruses, tumorigenic cell lines were similarly recovered. The co-infection of primary bone marrow cells seems to yield more transformed cell lines than does sequential addition of the same oncogenes to a cell line. The fact that the outgrowths from co-infection of a heterogeneous population were clonal, while those produced in sequential infection of a cell line were polyclonal may point towards an explanation. The clonal outgrowths of co-infection may represent the dominance of a rare and more highly transformed cell over many other cells carrying one or both oncogenes. The fact that only two of 20 cultures contained 109 co-infected clones and that their outgrth required 2 months may reflect the necessity for other oncogenic events. While lL-7 and v-Ha-ras expression may contribute to the transformed phenotype, the cooperation of other rare oncogenic events is critical to the tumorigenic phenotype. in the superinfection of1|12R4 cells, the more homogeneous population offers less opportunity for detecting rare transforming events. in addition, superinfected populations were recovered more rapidly (3 weeks). The sequential introduction of oncogenes into clonal cell lines seems to provide 6 more accurate assessment of their contribution to cooperative transformation than co-infection of a heterogeneous population. Materials and methods: Viruses. lL-7(SH) (Young et al., 1991) is derived from the Moloney murine sarcoma virus vector, pMV6tKneo (Kirschmeier et al., 1988). A cDNA for murine lL-7 that lacks translation-inhibiting flanking regions is expressed from the viral long terminal repeat (LTR), as well as the Tn5 neo gene (G418-resistant) from the herpesvirus thymidine kinase promoter. MMCV-neo (Wagner et al., 1985) is derived from Moloney murine leukemia virus (MoMuLV) and Harvey murine sarcoma virus. it expresses the v-myc gene of retrovirus OK10 from the viral LTR as a subgenomic mRNA and the Tn5 neo gene from the herpesvirus thymidine kinase promoter. SV(X)-Ha-ras (Schwartz et al., 19866) is derived from the MoMuLV vector, leP-NEOSV(X)1 (Cepko et al., 1984). it expresses v-Ha-ras from the viral LTR and the Tn5 neo gene as a subgenomic mRNA from the viral LTR. Virus stocks consisted of culture medium from NIH3T3 cell lines that had 110 been cotransfected with a proviral clone of MoMuLV and lL-7(SH), MMCV-neo, or SV(X)-Ha-ras. For the helper-free SV(X)-Ha-ras stock, culture medium was collected from the 1112 cell line (Mann et al., 1983) that had been transfected with SV(X)-Ha-ras. Virus titers of 2x105 to 8x105 G418-resistant colonies per ml were obtained for lL-7(SH); 2.4x106 for MMCV-neo; 1.0x105 to 1.5x105 for SV(X)-Ha-ras. Helper-free SV(X)-Ha-ras had a titer of 5x104 G418-resistant colonies per ml. Cell culture and viral Infections. Bone marrow from 3— to 5-week old BALB/c mice was cultured by the procedure of Whitlock and Witte (1982) with the addition of a viral infection step. Bone marrow was suspended at 2.0x10° cells per ml in RPMI 1640 medium supplemented with 5% fetal calf serum and 5x10"5 M 2-mercaptoethanol. To this was added an equal volume of virus stock and Poiybrene (Sigma Chemical Co., St Louis, MO) to a final concentration of 8 pg /mi. After 3 hours of incubation at 37° C, the cells were pelleted and resuspended at 106 cells per ml in RPMI 1640 supplemented with 5% fetal calf serum and 5x105 M 2 mercaptoethanol. 5 ml of this suspension was plated per 6-cm culture dish. 1|:2R4 cells were cultured in RPMI 1640 supplemented with 5% fetal calf serum and 5x10‘5 M 2-mercaptoethanoi over a feeder culture of adherent bone marrow cells (Whitlock et al., 1983). For superinfection, these cells were suspended in the same medium at 105 cells per ml and mixed with an equal volume of virus stock and Poiybrene as described above. All cultures were fed twice weekly with RPMI 1640 supplemented with 5% fetal calf serum and 5x10'5M 2-mercaptoethanol. Once a week approximately 80% of the spent medium was replaced with fresh medium. The cultures were expanded by transfer of nonadherent cells to feeder cultures (Whitlock et al., 111 1983). Growth in soft agar medium was performed over a feeder culture as described by Whitlock et al. (1983). Nucleic acid isolation and analysis. Cytoplasmic RNA was isolated by a sodium dodecyl sulfate-urea procedure as described by Schwartz et al. (1981). High molecular weight DNA was isolated from nuclei collected in the preceding procedure by a method described in Schwartz et al. (19866). Restriction enzyme-digested DNAs were electrophoresed through 0.8% agarose. RNAs were electrophoresed through 1% agarose-formaldehyde gels (Rave et al., 1979). Transfer and hybridizations were washed to a stringency of 0.1 x SSPE in 0.1% SDS. Hybridization probes were prepared by random priming using a kit from United States Biochemical Corp. (Cleveland, OH) with the incorporation of 5’-[a-32P]dATP (3,000 ci/mmol; lCN, Costa Mesa, CA). The lL-7 probe was a 0.5 kb Pstl fragment of the murine IL-7 cDNA (Young et al., 1991). The v-Ha-ras probe was a 0.46 kb EcoRI fragment corresponding to v-Ha-ras-encoding sequences of Harvey murine sarcoma virus (Ellis et al., 1980). The v-myc probe was a 0.8 kb CIal-BamHI fragment corresponding to 3’-terminal v-myc-encoding sequences of retrovirus OK10 (Hayflick et al., 1985). The mu heavy chain probe was a genomic 1.9 kb BamH1-EcoRl fragment, which corresponds to the JH2, JH3, and JH4 regions that are 5’ to the mu heavy chain constant region gene (Early et al., 1980). The kappa light chain probe was a genomic 0.48 kb Hpal-Bglll fragment extending from a point about 50 bp within the 5’- terminus of the kappa light chain constant region gene to the 3’-terminal poly(A) addition site (Seidman and Leder. 1978). The GAPDH probe was a 1.3 kb cDNA (Fort et al., 1985). 112 Tumor challenges. Cells were washed twice in RPMI 1640 and were then resuspended in the same at 8x106 cells per ml. BALB/c mice, 3 to 5 weeks old, were injected intraperitonealiy with 0.25 ml of the cellular suspension. Animals were observed for a maximum of 73 days post injection. Animals were sacrificed and autopsied when they became moribund or at 10 weeks. Tumors were verified as being derived from the challenging cell lines by Southern blot analysis of retroviral integration sites (data not shown). Acknowledgments: This work was supported by Grant CA45360 from the National Cancer Institute. We thank Dr. Owen Witte for helpful discussions and critical review of this manuscript. 1 13 References: Cepko, C.L, Roberts, B.E. & Mulligan, RC. (1984). Cell, 37, 1053-1062. Chang, J.M., Metcalf, D., Lang, R.A., Gonda, R.J. 81 Johnson, GR. (1989). Blood, 73, 1487-1497. Chen, S.-C., Redenius, D. & Schwartz, RC. (1991). Biochem. Biophys. Res. Commun., 178, 1343-1350. Early, P., Huang, H., Davis, M., Calame, K. 8: Hood, L (1980). Cell, 19, 981-992. Ellis, R.W., DeFeo, D., Maryak, J.M., Young, HA, Shih, T.Y., Chang, E.H., Lowy, DR. 81 Scolnick, EM. (1980). J. Virol., 36, 408-420. Fort, P., Marty, L., Piechaczyk, M., El Salrouty, S., Dani, C., Jeanteur, J. & Blanchard, J.M. (1985). Nucl. Acids Res, 13, 1431-1442. Hayfiick, J. Seeburg, P.H., Ohlsson, R., Pfeifer-Ohlsson, 8., Watson, 0., Papas, T. 81 Duesberg, PH. (1985). Proc. Natl. Acad. Sci. USA. 82, 2718-2722. Kirschmeier, P.T., Housey, G.M., Johnson, MD, Perkins, AS. 81 Weinstein, LB. (1988). DNA, 7, 219-225. Kremer, N. E., D’Arcangeio G., Thomas S.M., DeMarco M., Brugge J.S., and Halegoua S. (1991). J. Cell Biol. 115, 809-819. Mann, R., Mulligan, R.C. & Baltimore, D. (1983). Cell, 33, 153-159. Meeker, T.C., Hardy, D., Willman, C., Hogan, T. 8: Abrams, J. (1990). Blood, 76, 285-289. Morrissey, P.J., Goodwin, R.G., Nordan, R.P., Anderson, 0., Grabstein, KH., Cosman, 0, Sims. J., Lupton, 8., Acres, B., Reed, S.G., Mochizukl. D., Eisenman, J., Conion, P.J. 81 Namen, AE. (1989). J. Exp. Med., 169, 707-716. Muirhead, M., Davis, R., Schwartz, R.C., Waldschmidt, T.J., Ackerrnann, L, Palumbo, G. 8 Smith, R.G. (1990). intl. J. Cell Cloning, 8, 392—408. Namen, AE., Lupton, 8., Hjerrild, K., Wignall, J., Mochizukl. D.Y., Schmierer, A., Mosby, B., March, C.J., Urdal, D., Gillis, 8., Cosman, D. 8: Goodwin, RG. (1988). Nature, 333, 571-573. Overell, R.W., Clark, L, Lynch, 0., Jerzy, R., Schmierer, A., Weisser, KE., Namen, A.E. 81 Goodwin, R.G. (1991). Moi. Cell. Biol, 11, 1590-1597. Rave, N., Crkvenjakov, R. & Boedtker, H. (1979). Nucl. Acids Res, 6, 3559-3567. Sawyers, C.L, Denny, CT. 81 Witte, ON. (1991). Cell, 64, 337-350. Schwartz, R.C., Sonenshein, G.E., Bothweli, A. & Gefter, ML (1981). J. lmmunol., 126, 2104-2108. Schwartz, R.C., Stanton, LW., Riley, S.C., Marcu, KB. 81 Witte, O.N. (19866). Mol. Cell. Biol., 6, 3221-3231. Schwartz, R.C., Stanton, LW., Marcu, KB. 8: Witte, O.N. (1986b). Curr. Topics Micro. Immunol., 132, 75-80. Schwartz, R.C. & Witte, ON. (1988). Mutation Res., 195, 245-253. 1 14 Seidman, JG. 8: Leder. P. (1978). Nature, 276, 790-795. Wagner, E.F., Vanek, M. & Vennstrom, B. (1985). EMBO J., 4, 663-666. Whitlock, C.A. & Witte, ON. (1982). Proc. Natl. Acad. Sci. USA, 79, 3608-3612. Whitlock, C.A., Ziegler, S.F., Trieman, L.J., Stafford, J.|. & Witte, ON. (1983). Cell, 32, 903-911. Wong, P.M.C., Chung, S., Dunbar, C.E., Bodine, D.M., Ruscetti, SR. 8: Nelnhuis, AW. (1989). Mol. Cell. Biol., 9, 797-808. Young, J.C., Gishizky, M.L. & Witte, ON. (1991). Mol. Cell. Biol., 11, 854-863. APPENDICES APPENDIX A Lineage Switch Macrophages Can Present Antigen‘ ABSTRACT: Recent reports of "lineage switching" from a lymphoid to macrophage phenotype have left unresolved the question of whether such cells are functional macrophages or nonfunctional products of differentiation gone awry. This study demonstrates that several "macrophage-like" cell lines derived from v—Ha—ras-transformed pre-B cells have gained the capacity to effectively present antigen in an MHC-restricted fashion. Using an assay involving the co-cultivation of putative antigen-presenting cells with chicken ovalbumin (cOVA) and a cOVA-specific T cell hybridoma, "lineage switch” cell lines were found to present antigen as effectively as macrophage-containing peritoneal exudates. Neither the original pre-B cell precursors nor B cell lymphomas derived from them present antigen. Thus we have demonstrated that these “lineage switch" macrophages are capable of antigen presentation, a mature differentiated function. While gaining macrophage characteristics, these cells have also rearranged their kappa light chain immunoglobulin locus, suggesting that macrophage differentiation and immunoglobulin rearrangement are not mutually exclusive processes. The existence of both lymphoid and myeloid characteristics in a cell fully capable of antigen presentation suggests greater plasticity in hematopoietic lineage commitment than conventionally thought to be the case. 1James Bretz, Shu-Chih Chen, Diane Redenius, Hsun-Lang Chang, Walter J. Esseiman, and Richard Schwartz. Department of Microbiology, Michigan State University, East Lansing, Mi 48824- 1 101 115 1 16 INTRODUCTION: The concept that hematopoietic differentiation involves an early and irreversible lineage commitment is brought into question by numerous observations of leukemias and lymphomas that express myeloid or lymphoid markers outside of their respective lineages. The coexpression of differentiation markers has been interpreted as being either an aberrant phenomenon caused by leukemogenesis (McCulloch, 1983) or a reflection of the normal but transient existence of bipotentiai progenitors in hematopoiesis (Greaves et al., 1986). in particular, the existence of a number of transformed cell lines with both lymphoid and macrophage characteristics has suggested a close relationship between these lineages. Murine macrophage cell lines have been derived from lymphoid tumors and from in vitro transformants induced either by murine leukemia viruses or chemical carcinogens (Boyd and Schrader, 1982; Holmes et al., 1986; Hanecak et al., 1989). Three groups have studied systems where a transition from a lymphoid to a macrophage phenotype could be induced. Klinken et al. (1988) demonstrated that B lymphoid cells from transgenic mice that express c—myc using the immunoglobulin mu enhancer could be induced to take on macrophage-like characteristics when infected with a retrovirus expressing v—raf. Davidson et al. (1988) showed that a v—Ha—ras transformed lymphoid cell line could be stimulated by Iipopolysaccharide (LPS) to differentiate along either the lymphoid pathway into pre—B-like cells or along the myeloid pathway into macrophage-like cells. Recently, Borzillo et al. (1990) reported the CSF—1 dependent macrophage lineage transition of a pre—B cell line expressing the human CSi‘—1 receptor. The macrophage-like cell lines that have been derived from B lymphoid cells 117 have been classified as macrophage on the basis of their morphology, expression of MAO—1, MAC—2, a-naphthyi acetate esterase and iysozyme, and their ability to phagocytose latex beads. More functional assays for antigen presentation and tumoricidal activity that would establish whether these cells could act in vivo similarly to authentic macrophages have not been presented. in this paper, we demonstrate the ability of several macrophage cell lines derived from v—Ha—ras-transformed pre—B cells to present antigen to a T helper cell hybridoma. RESULTS: A tumor consisting of adherent cells with a macrophage morphology was identified during our studies on the tumor progression of a pre-B lymphoid cell line expressing v-Ha-ras (Chen et al., 1991). This tumor, designated tumor 4, was derived from a clonal cell line, designated R2, that was generated by infection of fresh murlne bone marrow with a mixture of a v-Ha-ras-expressing retrovirus and Moloney murine leukemia virus (MoMuLV) (Schwartz et al., 1986a). The R2 cell line was classified as being a pre-B cell on the basis of several criteria. It possessed 6 blast cell morphology with a large nucleus and scant cytoplasm. it expressed the B lineage—specific marker, B220 (Coffman and Weissman, 1981). While not expressing detectable immunoglobulin mu chain, R2 showed a rearrangement in the DNA of that locus. The immunoglobulin kappa chain locus was in a germiine configuration. Tumor 4 is Derived from the R2 Cell IJne. in order to ascertain whether we had identified a probable instance of 118 Figure 1. Viral integrations. Southern blot analysis of DNAs from liver (L), R2 and tumor 4 (T4). (A) DNA was digested with EcoRI and 10 pg of each sample was electrophoresed through 0.8% agarose. The blot was probed for v—Ha—ras. (B) DNA was digested with Bglll. The blot was probed for murine ecotropic env sequences. Size markers are the positions of an ethidium bromide-stained Hindill digest of bacteriophage A and are denoted in kilobases. 119 lineage switching, it was necessary to demonstrate that tumor 4 was derived from R2. To that end, the sites of integration of the v-Ha-ras-expressing retrovirus and MoMuLV were compared between the tumor and the cell line. Southern hybridization analysis of EcoRI-digested DNA with a v-Ha-ras probe showed that tumor 4 contained the same 5.3 kb proviral integration fragment as R2 (Figure 1A). This proviral integration fragment is defined by a 3’ EcoRI site internal to the viral genome and a 5’ EcoRI site peculiar to the site of integration. In addition to the 5.3 kb fragment, there is a 23 kb fragment representing the endogenous c-Ha-ras in all the DNAs. Southern hybridization analysis of Bglll-digested DNA, using a probe for the ecotropic MuLV env gene, revealed similar MoMuLV integration fragments in R2 and tumor 4 (Figure 1B). The MoMuLV genome possesses a Bglll site within env, such that the above hybridization would detect a fragment extending from that Bglll site to a Bglll site in the host cell genome flanking the 3’ terminus of the provirus. These data demonstrate that the putative macrophage tumor was derived from the pre-B cell line. Tumor 4 Cells Possess Macrophage Characteristics. Tumor 4 was initially suspected to be a macrophage because of the large size of its cells and its adherent growth in cell culture. Microscopic examination of Wright-Glemsa stained cells confirmed their large size and revealed the cells of tumor 4 (Figure 28) to have a much more extensive and granular cytoplasm than R2 (Figure 2A). An immunoperoxidase detection procedure found tumor 4 cells to have retained some expression of B220, and to have gained expression of high levels of MAC-1 (data not shown). MAC-1 is generally considered to be a marker Figure 2. Nonspecific phagocytosis of latex beads. The cells were Wright-Glemsa stained and photographed at 200x magnification. (A) R2; (B) tumor 4. 121 for cells of the myeloid lineage (Springer et al., 1979). Histochemical proceduresrevealed a high level of a-naphthyl acetate esterase activity in tumor 4 cells, which is not found in R2 cells (data not shown). This is an enzyme activity generally associated with cells of the monocyte-macrophage lineage (Rogers et al., 1980). Tumor 4 cells (Figure 2B) were positive for the nonspecific phagocytosis of latex beads, while R2 cells (Figure 2A) were not. Nonspecific phagocytosis is another marker of the monocyte-macrophage lineage (Raschke et al., 1978). These data strongly suggest a macrophage phenotype for tumor 4 cells. At late stages of myeloid differentiation, the levels of c-myc and c-myb mRNA decrease, while the level of c-fms mRNA increases (Gonda and Metcalf, 1984; Sheng-Ong et al., 1987). The levels of mRNA from these protooncogenes detected in tumor 4 cells was consistent with tumor 4 having advanced to a late stage of myeloid differentiation. Cytoplasmic poly A+ RNAs of the parental R2 cell line, tumor 4 and six other tumors derived from R2 that had lymphoid characteristics were examined by Northern hybridization analysis (Figure 3). One blot was hybridized successively with c-myc and fl 2-microglobulin probes. Another blot was hybridized successively with c-myb, c-fms and rat gcheraldehyde-B-phosphate dehydrogenase (GAPDH) probes. Hybridization to the fi2-microglobulin and GAPDH probes provided a control for gel loading. Tumor 4 cells clearly show reduced levels of c-myc and c-myb expression in comparison to R2 cells and lymphoid tumors. in contrast c-fms expression is elevated in tumor 4 cells. A cell line with macrophage characteristics has also been isolated from tumor 5 cells, which show elevated c-fms expression (Figure 3). 122 c-Mvc~ I! 1’1 17 11 1" 1'5 1'! ‘7 ”fl” 9.5 p m nannn'nnnn Deals-rel.” Figure 3. RNA analyses of c—myc, c—myb and c—fms. Northern blot analyses were performed on polyA+ RNA from R2 and seven tumors (T 1—17). Each sample of RNA was the polyA+ fraction selected from 150 pg of total cytoplasmic RNA. One blot (upper panel) was probed successively for both c-myc and BZ-microgiobulin, while the other blot (lower panel) was probed successively for c—myb, c—fms and rGAPDH. 123 Another aspect of macrophage function is the ability to release cytokines in response to LPS stimulation. We examined the presence of iL-1, lL—6 and TNF in the media of cells cultured in the presence or absence of 10 pg/ml of LPS for 24 hours. Cellular proliferation assays for lL—1 and lL-6, and a cytotoxicity assay for TNF revealed varying levels of cytokine release for six subclones of tumor 4 (see below), while the parental R2 pre—B cell line did not elaborate any of these cytokines except low levels of lL-1 (Table 1). The LPS-inducible release of cytokines was again consistent with a macrophage phenotype for tumor 4 cells. Tumor 4 Cells Also Show Differentiated Lymphoid Characteristics. Davidson et. al. (1988) found that a v-Ha-ras-tranformed lymphoid cell line could be stimulated to differentiate along either the myeloid or lymphoid pathways. Since tumor 4 cells showed a variety of DNA rearrangements in the kappa light chain locus (data not shown), it was of interest to determine whether the cells that had gone on to rearrange the kappa locus were the same cells that had progressed toward a macr0phage phenotype or whether the tumor 4 cells were a mixed population of B cells and macrophages. To that end, tumor 4 cells were plated in soft agar medium and six subclones were recovered. Southern hybridization analysis of EcoRI-digested DNA isolated from the subclones showed that they all contained the same 5.3 kb v-Ha-ras proviral integration fragment as R2 and tumor 4 cells (data not shown; see Figure 1A). The six subclones of tumor 4 possessed the same myeloid characteristics described above for the uncloned tumor, but varied in their pattern of kappa light chain gene rearrangement. Southern hybridization analysis of BamHl-digested DNAs with a kappa probe revealed that subclones 3 and 5 possessed one germiine and one rearranged kappa allele, while subclones 124 Table 1. LPS-induced cytokine release by tumor 4 macrophage subclones. lL-1 (U/ML) IL-6 (U/ML) TNF (U/ML) -LPS +LPS -LPS +LPS -LPS +LPS r12 0 2 0 0 0 0 T4.1 0 2 0 1 0 0 74.2 0 6 0 100 0 40 T43 0 6 0 1500 0 65 74.4 0 19 0 39 0 34 T45 1 4 0 0 0 0 T4.6 o 4 0 0 o 0 Table 1. The capacity of cell lines to release the cytokines lL—1, IL—6 and tumor necrosis factor (T NF) was determined by assaying culture supernatants. For this purpose, cell lines were incubated for 24 hours at 2.5x10‘5 cells/mi with 10 pg/ml LPS in RPMI 1640 supplemented with 10% fetal calf serum and 5x10’5 M 2-mercaptoethanol. Culture supernatants were collected, passed through a 0.2 micron filter and stored 6t—70° C until assayed. lL—1 activity was assayed by its ability to induce proliferation of D10.G4.1 cells in the presence of Concanavalin A as described by Ayala et al. (19906). lL-6 activity was determined by its ability to induce the proliferation of the 7TD1 B-cell hybridoma as previously described by Huitner at al. (1989). TNF activity was assessed by its cytotoxicity to WEHl—164 clone 13 cells as previously described by Ayala et al. (1990b). The relative units of cytokine activity were determined by comparison of the activity of dilution series of experimental supernatants to the activities of dilution series of purified human lL—1 (Genzyme), recombinant human lL—6 (Amgen Corp.) or murlne TNF—alpha (Amgen Corp.) standards. 125 xiii»: ’5‘" .A .51 14.1 . v (‘0 E 3% 5% 2’; Flgure 4. Kappa light chain rearrangements. Southern blot analysis of DNAs from liver (L), R2 and six subclones of tumor 4 (1—6). DNA was digested with BamHI and 10 pg of each sample was electrophoresed through 0.8% agarose. The blot was probed for kappa light chain constant region sequences. Size markers are the positions of an ethidium bromide-stained Hindill digest of bacteriophage A and are denoted in kilobases. 126 1,2,4 and 6 possessed rearrangements in both alleles (Figure 4). All the subclones possessed a rearranged BamHI fragment of approximately 7 kb. Tumor 4 was apparently derived from an outgrth of R2 that had undergone this rearrangement. Some of the subclones then proceeded to rearrange their other kappa allele. Clearly, tumor 4 contained cells which individually had differentiated along both the lymphoid and myeloid pathways. Having observed kappa light chain rearrangements in macrophage subclones of tumor 4, we next examined the status of immunoglobulin expression by Northern blot analysis. Kappa light chain transcript could not be detected (data not shown). A mu heavy chain probe revealed a diverse range of RNAs in the macrophage subclones (Figure 5) that correspond in size to 1.9, 2.1, 2.3 and 2.9 kb transcripts reported to be initiated in the mu switch region of myeloid cell lines (Kemp et al., 1980). The R2 pre-B cell line possesses predominantly larger RNA species that include those that correspond in size to mature mu mRNAs of 2.4 and 2.7 kb. These species are diminished upon lineage switch. Comparison to a hybridization of the same blot with a probe for GAPDH (Figure 5) shows the R2 RNA to be underloaded and thus the diminution of mu transcription in the macrophages is even more dramatic than apparent from casual inspection of the data. Apparently, the macrophage subclones of tumor 4 lose the capacity to transcribe functional mu mRNA, even though the rearrangement of the kappa locus suggests progress in lymphoid differentiation. Since CD45 isoforms have been reported to be lineage specific (Saga et al., 1987; Streuli et al., 1987; Ralph et al., 1987), the expression of this surface marker was examined among the subclones of tumor 4. An immunoperoxidase detection 127 MU 14 Melon“ 221234.16 Figure 5. Expression of mu heavy chain RNA. Northern blot analysis was performed on poly A+ RNA from R2 and six subclones of tumor 4 (1-6). Each sample of RNA was the poly A+ fraction selected from 100 pg of total cytoplasmic RNA. The blot was probed for mu heavy chain. The positions of ethidium bromide-stained rRNAs are noted on the right. The positions of mu RNA species are marked on the left and denoted in kilobases. The lower panel shows the same blot probed for GAPDH as a control for loading. 128 procedure detected 8220, the B lymphoid isoform of CD45, in tumor 4 cells. The expression of CD45 was further examined among the tumor 4 subclones in orderto determine the relative expression of the B220 isoform in comparison to the isoform that predominates in myeloid cells. Recently, Chang et. al. (1989) described the use of a reverse transcription-polymerase chain reaction (RT-PCR) technique to determine the pattern of alternate exon use in CD45 expression of hematopoietic cells. They found that B lymphoid cell lines uniquely expressed a form of CD45 mRNA possessing 3 optional exons, while two myeloid cell lines (a macrophage and a mast cell) predominantly expressed a form lacking these exons. We utilized RT—PCR to examine CD45 expression among R2 and the subclones of tumor 4 (Figure 6). All of the cell lines expressed multiple species of CD45 mRNA. Subclones 2,3,4 and 6 expressed a CD45 mRNA containing 3 optional exons, typical of B lymphoid cells, while subclones 1 and 5 predominantly expressed mRNA lacking these exons, typical of myeloid cells. R2 expressed the expected three exon 8 lymphoid isoform. Thus the pattern of CD45 expression is heterogeneous among macrophage subclones of the same tumor. The subclones of tumor 4 can function effectively in antigen presentation. In order to test the ability of the tumor 4 subclones to present antigen, an assay system required an antigen-specific T helper cell line that could be stimulated to produce interleukin 2 (IL-2) upon presentation. For these experiments, the putative antigen-presenting cells were co-cultivated with a T cell hybridoma specific for chicken ovalbumin (cOVA) and restricted for l—Ad (the haplotype for BABL/c), DO.11.10/54.4. In the presence of cOVA, authentic macrophages such as those in a peritoneal exudate stimulate the hybridoma to produce lL-2 (Figure 7A). IL-2 129 production was assayed by the application of media supernatants from co-cultivations to an IL—2 dependent cell line, CTLL-2. All of the macrophage-like tumor 4 subclones displayed antigen presentation capacities comparable to peritoneal exudates (Figure 7A). Furthermore, all of the tumor 4 subclones showed antigen presentation capacities dramatically greater than either the parental R2 pre-B cell line or tumor 1, a B cell tumor derived from R2 (Figure 7A). lL-2 production was dependent on the presence of cOVA during co-cultivation of presenting cells with cells of the helper T cell hybridoma. Supematants produced in the absence of cOVA were analyzed for all the cell lines and the values for lL—2 production were found to be near zero (data not shown). These control values were subtracted from those determined for supernatants produced in the presence of cOVA to generate the data presented in Figure 7A, B and C. lL—2 production was also dependent on the presence of T cell hybridoma cells. Supematants produced by incubations of putative presenting cells with cOVA in the absence of T cell hybridoma cells had no detectable lL—2 (data not shown). The ability to present antigen was not stimulated by exposure to LPS for any of these cell lines (data not shown). A macrophage-like outgrth from tumor 5 (also derived from R2) and the cells of a macrophage-like tumor derived from the pre-B cell line R1 (9) showed levels of antigen presentation similar to those observed for the tumor 4 subclones (Figure 7B). The ability to present antigen may therefore be a common phenomenon among v-Ha-ras-transformed B lymphoid cells that acquire macrophage-like characteristics. Authentic antigen presentation should be MHC—restricted, so all of the putative antigen-presenting cells were also co-cultivated with a T cell hybridoma 130 specific for cOVA and restricted for l—Aq, 30023-244. As exemplified by subclone 4 of tumor 4 (T 4.4) and the macrophage tumor derived from R1 (R1T), the antigen presentation observed is MHC-restricted (Figure 7C). l-A Expresslon Antigen presentation to T cells requires la expression and the observation of l—Ad-restricted presentation (Figure 70) indicates that these "lineage switch" macrophages express l—Ad. In order to assess whether the acquisition of presentation capacity correlated with acquisition of la expression, in particular l—Ad, we performed flow cytometry with FITC-conjugated anti-mouse l-Ad on the macrophage cell lines and their pre—B cell precursors. While both R1 and R2 (pre-B cells) displayed no detectable l—Ad, the macrophage cell lines represented by R1T and T44 showed a low expression of l—Ad (Figure 8). 131 Figure 6. RT—PCR analysis of CD45. RT—PCR was performed on the polyA+ RNAs of R2, the six subclones of tumor 4 and P388D1 (a myeloid control). The products were electrophoresed through 2% agarose and stained with ethidium bromide. (C) 3-exon plasmid control; (1—6) T4 subclones; (M) 123 bp ladder; (Mp) P388D. PCR products smaller than the 0 exon product may represent an RNA species lacking an additional exon (Chang and Esselman, unpublished results). 132 '. _. - . 1 .\ o :93 B W ; 0 Ti : ~ Q i o T41 E .. ‘. $5111 3 v ‘1“: 3 H“ LJ g v T4 3 E 9 .1 a T4 4 3 p 7: I T4 -~ '? - '5 A T4 0 E: ‘2 '3 e g E - L"! 3) l‘ f‘ 4 l 1' T V l 3 . 1 1 v . O 2 4 6 3 'C ‘; ’4 ‘6. '5 ;I‘ 2: G 2 4 6 8 “O '2 34 i6 ‘8 23 4 No. of presenting cells/culture ix 10 i No. of presenting cells/culture (x 10‘) ( v 6 .. \ V 'AT ”-31 v RlT ll-A“) 0 14411-4 l o T14 ll-qu "-2 produced (units/ml) (A I i . 1 I T Tr T O l 2 3 4 5 5 7 8 9‘ 10 No. of presenting cells/culture ix ii) i Figure 7. Antigen presentation. (A) Antigen presentation assays were performed on R2, the tumor 4 subclones (4.1—4.6), tumor 1 (a B cell tumor derived from R2), and peritoneal exudates (er). (B) Antigen presentation assays were performed on R2, a macrophage outgrth of tumor 5 (TS; derived from R2 in a different animal), R1 (another pre-B cell line transformed by v-Ha-ras). and R1T (a macrophage tumor derived from R1). (C) Antigen presentation assays were performed on a tumor 4 subclone (T 4.4) and R1T with DO-11.10/54.4 (l-A'd-restricted) or 30023-244 (l-Aq-restricted). Each point represents an average value obtained from a dilution series for each presentation supernatant, testing the response of CTLL-2 cells to lL—2 in those superantants. The results shown are representative of at least two experiments with each cell line. 133 Allliiiflliifi 4’ Joacogufioo >332; 0... .823 P... 328.2093 .6 33a of ~¢ ace 2. .8; 43.: v2.2.3 2.3.6.: 33: canon—580-92“. Jobcoo o no Canoe: u_.o..u<-_._sc. 33.24.3090»: 5:. 3033923: 2». one «:2 .3. .2. co Eaton mo: F528;“. so: mozmomNEODJu F. _ _ P . . mi _ PE pm — P _ p b n _ «.3. «I _ £339.98 31. .o v.53... SLNDOO 134 DISCUSSION: This study demonstrates the capacity of several macrophage-like tumor cell lines derived from v—Ha—ras-transformed pre—B cell lines to present antigen with MHC-restriction. This finding establishes that cells having undergone "lineage switching" can perform a function normally associated with a fully differentiated macrophage or B cell. While numerous examples exist of B lymphomas with the capacity to present antigen (Chesnut et al., 1982; Walker et al., 1982), neither the pre—B cell precursors of the macrophage-like cell lines nor B cell lymphoma cell lines derived from those precursors could present antigen. Thus the capacity to present antigen appears to correlate with the differentiation of these cells along the macrophage lineage. Indeed, two cell lines with the most dramatic level of antigen presentation had lost expression of the B cell isoform of CD45 and displayed a pattern of CD45 more typical of a myeloid cell (subclones 1 and 5, Figure 6; T4.1 and T45, Figure 7A). Perhaps, loss of the B cell isoform of CD45 is indicative of further maturation along the myeloid lineage. it may be worthwhile to investigate the role of CD45 in macrophage function. The fact that similar antigen presentation abilities were found in macrophage derivatives of two completely independent cell lines (R1 and R2) suggests the generality of this phenomenon. Since it is well established that lL-1 along with antigen presentation is an important co-activator of T cells, it is surprising that the inducibility of cytokine release by LPS (Table 1) does not correlate with the effectiveness of antigen presentation by the T4 subclones (Figure 7A). Apparently the low levels of lL—1 that some of these macrophages are capable of elaborating is sufficient for T cell activation. The observation that two of the best lines for antigen presentation 0' 4.1 135 and T45) have a weak response to LPS suggests that LPS-induced cytokine release may not be an adequate measure in itself for evaluating macrophage function. The "lineage switch" macrophages reported here express a low level of la (Figure 8). This is consistent with the previous report of Davidson et al. (1988). The precursor pre—B cells lack detectable la. Perhaps la expression is the critical property determining the capacity to present antigen among these cells. Certainly, la expression is necessary for antigen presentation, but its sufficiency for antigen presentation among the cell lines we have studied will require further experimentation. The six macrophage-like subclones of tumor 4, while possessing a common rearranged kappa allele, displayed a variety of kappa light chain gene rearrangements at their other kappa allele. Compared to their parental cell line, these cells have progressed along the B as well as the monocyte/macrophage lineage. The varying rearrangements of one kappa allele suggests rearrangement subsequent to macrophage conversion and that at least certain elements of lymphoid and macrophage differentiation programs are not mutually exclusive. The fact that the R2 cell line can also generate a lymphoma (T 1, Figure 7A) that expresses both mu and kappa chains (data not shown) demonstrates the potential of this cell line to differentiate quite far along either the lymphoid or macrophage pathways. The relationship between lymphoid and macrophage differentiation revealed in these cells differs somewhat from that seen in cases of "lineage switch“ previously reported. Klinken et al. (1988) found “lineage switch“ macrophages at both the pre-B and B cell stages of immunoglobulin rearrangement. However, they 136 did not find macrophages that had progressed in their immunoglobulin rearrangement compared to their lymphoid cell precursors, as we have. Davidson et al. (1988), examining v—Ha—ras-transformants similar to those reported here, could induce those cells to differentiate into either lymphoid or macrophage cells upon exposure to LPS. The lymphoid derivatives they reported did not progress beyond the pre-B cell stage, while we have identified an immunoglobulin producing tumor derived from a pre-B cell line that also gave rise to a macrophage tumor. Perhaps the more complex environment provided during tumor challenge allowed the cells described here to more fully develop along the lymphoid lineage when that pathway was selected. At any rate, the v—Ha—ras-transformed pre-B cells described here seem truly bipotentiai. The ability of these cells which undergo an apparent "lineage switch" to perform a fully differentiated function presents the possibility that they may represent an unusual but normal subset of hematopoietic cells rather than an oddity induced by transformation. The existence of both lymphoid and macrophage characteristics in a cell fully capable of antigen presentation suggests greater plasticity in hematopoietic lineage commitment than conventionally thought to be the case. MATERIALS AND METHODS: Cell Lines. R1 and R2 are v—Ha—ras-transformed murine pre—B cell lines described in Schwartz et al. (1986a). Tumors derived from R1 and R2 were generated as described in Schwartz et al. (19863,b) in syngeneic BALB/c mice and in BALB/c athymic nude mice. Briefly, cells were washed twice in RPMI 1640 and 137 were then resuspended in the same at 8x106 cells per ml. Five week old mice were injected intraperitoneally with 0.25 ml of the cellular suspension. Tumor 4, in particular, was isolated from an inguinal lymph node at 74 days post injection. Tumor cell lines were readily produced from explanted tumors by dispersal and transfer to feeder cultures of adherent bone marrow cells (Whitlock et al., 1983). All of these cell lines were cultured over feeder cells in RPMI1640 supplemented with 5% fetal calf serum and 5x10'5 M 2-mercaptoethanol. The subclones of tumor 4 were generated from single colonies grown in soft agar medium as described by Whitlock et al. (1983). The T cell hybridoma, DO—11.10/54.4, was a generous gift of Drs. Philippa Marrack and John Kappler (University of Colorado, Denver) (White et al., 1983). This hybridoma is specific for chicken ovalbumin in the context of l—Ad and crossreacts weakly with chicken ovalbumin in the context of l—Ab. 30023-244, another T cell hybridoma, was also a gift of Drs. Marrack and Kappler. This hybridoma is specific for chicken ovalbumin in the context of either l-Aq or l—E. CTLL-2 is a T cell line responsive to lL—2 and was obtained from the ATCC. All of these cell lines were cultured in RPMI1640 supplemented with 10% fetal calf serum and 5x10"5 M 2-mercaptoethanol in the absence of any feeder cells. Peritoneal exudates containing macrophages were produced from BALB/c mice treated 1 week previously with a 0.5 ml intraperitoneal injection of pristane. Nucleic Acid Analysis. Cytoplasmic RNA was isolated from actively growing cells by a sodium dodecyl sulfate-urea procedure as described by Schwartz et al. (1981). Poly A+ RNA was selected by oligo—dT cellulose chromatography (Rave et al., 1979). RNA was denatured, electrophoresed in a formaldehyde-1% agarose 138 gel (15), and transferred to Nytran (Schleicher and Schuell) (Thomas, 1980). High molecular weight DNA was isolated from nuclei collected in the preceding RNA isolation procedure as described in Schwartz et al. (1986a). DNA was digested with restriction enzymes as noted in the figure legends, electrophoresed through 0.8% agarose and transferred to Nytran (Southern, 1975). Hybridization probes were prepared by nick translation (Rigby et al., 1979) through the incorporation of [a-32P] dATP (3000 Ci/mmol; lCN). The v—Ha—ras probe was the replicative form of phage M13mp10 containing a 0.46 kb EcoRI fragment corresponding to v—Ha—ras encoding sequences (Ellis et al., 1980). The env probe was a 0.8 kb BamHI fragment from the env region of Friend murlne leukemia virus and is specific for the env sequences of murine ecotropic retroviruses (Silver and Kozak, 1986). The rmyc probe was the 4.7 kb genomic Hindill fragment of murine emyc (Stanton et al., 1984). The murine c—myb probe was a cloned 2.4 kb cDNA (a generous gift of Dr. Timothy Bender, University of Virginia, Charlottesville). The fms probe was a cloned 2.7 kb CIal-BamHl fragment of the McDonough strain of feline sarcoma virus (Donner et al., 1982). The murine BZ-microglobulin probe was a cloned 0.5 kb cDNA (Parnes et al., 1981). The rat glyceraldehyde-S-phosphate dehydrogenase (GAPDH) probe was a cloned 1.3 kb cDNA (Fort et al., 1985). The murine kappa light chain probe was the replicative form of phage M13mp10 containing a genomic 0.48 kb Hpal-Bglll fragment extending from a point about 50 base pairs within the 5’ terminus of the kappa light chain constant region gene to the poly A addition site (Seidman and Leder, 1978). The murlne mu heavy chain probe was a cloned cDNA (p 12) which extends from CH2 to the 3’-untranslated region of the secreted form of mu mRNA (Rogers et al., 139 1980). All hybridizations were performed under aqueous conditions in 5 x SSC at 65° C and washed to a stringency of 0.1 x SSC at 65° C. Reverse Transcription-Palymerase Chain Reaction (RT-PCR). RT-PCR was performed according to the procedure of Chang et al. (1989; 1991) using poly A+ RNA as substrate. The primers were a sense primer specific to exon 2 (GCCCTTCTGGACACAGAAGT; base positions 167—186) and an anti-sense primer specific to exon 9 (AATTCACAGTAATGTTCCCAAACAT; base positions 764- 740) of the cDNA of murine CD45 (Thomas et al., 1987). cDNA was prepared by incubating 1 ug of poly A+ RNA for 60 min at 37° C with 200 units of MoMuLV reverse transcriptase in a 20 ul reaction volume containing 50 mM Tris-HCl (pH 8.3), 75 mM KCI, 3 mM MgCl2, 5 mM DTT, 100 149/ml BSA, 40 units RNasin, 500 uM dNTP and 200 ng of anti-sense primer. A 5 pl aliquot was used directly for PCR amplification in a 50 pl reaction volume containing 50 mM KCl, 10 mM Tris-HCl (pH 9.3), 3 mM MgCl2, 0.1% w/v gelatin, 500 uM dNTP, 400 ng of sense and anti-sense primers and 2.5 units of Taq polymerase. PCR was performed in a DNA Thermal Cycler (Perkin-Elmer-Cetus, Inc.) for 24 cycles. Each cycle consisted of 40 s at 94° C for denaturation, 15 s at 55° C for annealing and 30 s at 72° C for elongation. The first cycle was preceded by a 5 min incubation at 94° C and the last cycle followed by a 4 min incubation at 72° C. Cytological Analyses. Cells were cytocentrifuged onto a microscope slide and allowed to air dry overnight. The cells were then incubated with either rat anti-B220 (monoclonal 14.8) or rat anti-MAC—1 (Boehringer Mannheim). Goat anti-rat immunoglobulin-horseradish peroxidase (Boehringer Mannheim) was used in a secondary incubation for detection. The presence of a-naphthyl acetate 140 esterase was determined by cytochemical staining (Yam et al., 1971) with a Sigma research kit. Nonspecific phagocytosis of latex beads was assayed by the method of Raschke et al. (Raschke et al., 1978). Antigen Presentation. Assays for antigen presentation were performed in a manner similar to that described by Marrack et al. (1989). Briefly, the cell lines to be assayed for antigen presentation were titrated into 200 pl microcultures containing 105 cells of either the T cell hybridoma DO-11.10/54.4 or 30023-244, both of which produces lL—2 in response to the presentation of chicken ovalbumin (cOVA) in the context of l—Ad or i—Aq, respectively. These assays were carried out in RPMI 1640 supplemented with 10% fetal calf serum, 5x10'5 M 2-mercaptoethanol and, where required, cOVA at 1 mg/ml. After 24 hours, incubation supernatants from these cultures were assayed for IL-2 using CTLL-2, an lL—2—dependent cytotoxic T cell line. Two-fold serial dilutions of supernatants were added to 5x103 CTLL—2 cells in 100 pl microcultures and incubated for 48 hours at 37° C. MT‘I’ (Sigma), a substrate for production of a colored product indicative of cell survival (Mosmann, 1983), was added at 0.5 mg/ml and the cultures incubated for an additional 4 hours at 37° C. Acid-isopropanol (40 mM HCI) was then added to dissolve the MIT formazan reaction product. The optical density of each well was quantitated by an ELISA reader at a wavelength of 540 nm. The specific activity of IL-2 in the supernatants was determined by comparison to a standard curve produced through the use of purified recombinant lL—2 (Cetus lnc.). Flow Cytometry. Cells were stained in PBS, 2% FCS with either FlTC-conjugated monoclonal antibody AMS-32.1 (anti-mouse l-Ad) (Phar Mingen) or 141 FlTC-conjugated mouse lgGZb, K (Phar Mingen) as an isotype-matched control. Cells were then fixed in PBS, 2% FCS, 0.5% formaldehyde and stored at 4° C until analysis. Flow cytometry was performed using an Ortho Diagnostics Cytofluorograph 50-H. ACKNOWLEDGMENTS: This work was supported by Public Health Service grants CA45360 (ROS) and GM35774 (WJE) and by a grant from the Elsa U. Pardee Foundation (RCS). We are indebted to Alfred Ayala for the cytokine assays. The authors thank Donna Paulnock for helpful discussions, and Susan Conrad and Michele Fluck for thoughtful comments on this manuscript. ‘ 142 REFERENCES: 1. Avlv, H. and Leder, P. (1975) Purification of biologically active globulin messenger RNA by chromatography on oligothymidylic acid-cellulose. Proc. Natl. Acad. Sci. USA 69, 1408-1412. 2. Ayala, A., Perrin, M.M., Wagner, MA. and Chaudry, l.H. (1990a) Enhanced susceptibility to sepsis following simple hemorrhage: depression of Fc and 03b receptor-mediated phagocytosis. Arch. Surg. 125:70-75. 3. Ayala, A., Perrin, M.M., Meldrum, D.R., Enel, W. and Chaudry, l.H. (1990b) Hemorrhage induces an increase in serum TNF which is not associated with elevated levels of endotoxln. Cytoklne 3, 170-175. 4. Borzillo, G.V., Ashmun, RA and Sherr, C.J. (1990) Macrophage lineage switching of murine early pre-B lymphoid cells expressing transduced fms genes. Mol. Cell. Biol. 10, 2703-2714. 5. Boyd, AW. and Schrader, J.W. (1982) Derivation of macrophage-like lines from the pre—B lymphoma ABLS 8.1 using 5-azacytldine. Nature 297, 691-693. 6. Chang, H.L, Zaroukian, M.H. and Esselman, W.J. (1989) T200 alternate exon use in murlne lymphoid cells determined by reverse transcription-polymerase chain reaction. J. lmmunol. 143, 315—321. 7. Chang, H.L, Lefrancois, L., Zaroukian, M.H. and Esselman, W.J. (1991) Developmental expression of CD45 alternate axons in murlne T cells. Evidence of additional exon use. J. Immunol. 147, 1687-1693. 8. Chen, S.-C., Redenius, D. and Schwartz, RC. (1991) Tumorigenesis of a v-Ha-ras—expresslng pre-B cell line selects for c-myc activation. Biochem. Biophys. Res. Comm. 178, 1343-1350. 9. Chesnut, R.W., Colon, SM. and Grey, HM. (1982) Antigen presentation by normal 8 cells, B cell tumors and macrophages: functional and biochemical comparison. J. Immunol. 128, 1764-1768. 10. Coffman, R.L, and Weissman, LL. (1981) A monoclonal antibody that recognizes B cells and 8 cell precursors in mice. J. Exp. Med. 153, 269-279. 11. Davidson, W.F., Pierce, J.H., Rudikoff, S. and Morse, H.C. (1988) Relationships between B cell and myeloid differentiation. J. Exp. Med. 168. 389-407. 12. Donner, L, Fedele, LA., Garon, C.F., Anderson, SJ. and Sherr, G.J. (1982) McDonough feline sarcoma virus: characterization of the moiecularly cloned provirus and its feline oncogene (v-fms). J. Virol. 41, 489-500. 13. Ellis, R.W., De Feo, D., Maryak, J.M., Young, HA, Shih, T.Y.. Chang, E.H., Lowy, DR. and Scolnick, EM. (1980) Dual evolutionary origin for the rat genomic genetic sequences of Harvey murlne sarcoma virus. J. Virol. 36, 408-420. 14. Fort, P., Marty, L, Piechaczyk, M., El Salrouty, S., Dani, C., Jeanteur, J. and Blanchard, J.M. 1985. Various rat adult tissues express only one major mRNA species from the glyceraldehyde-S-phosphate dehydrogenase multlgenlc family. Nucl. Acids Res. 13, 1431-1442. 15. Gonda, T.J., and Metcalf, D. (1984) Expression of myb, myc and fos proto—oncogenes during the differentiation of murlne myeloid cells. nature 310, 249-251. 143 16. Greaves, M.F., Chan, LC., Furley, A.J.W., Watt, SM. and Molgard, H.W. (1986) Lineage promiscuity in hemapoletlc differentiation and leukemia. Blood 67, 1—11. 17. Hanecak, R., Zovich, D.G., Pattengale, PK and Fan, H. (19%) Differentiation In vino of a leukemia virus-induced B-cell lymphoma into macrophages. Mol. Cell. Biol. 9, 2264-2268. 18. Holmes, K.L., Pierce, J.H., Davidson, W.F. and Morse, H.C. (1986) Murine hematopoietic cells with pre-B or pre— B/myeloid characteristics are generated by in vitro transformation with retroviruses containing fes, ras. abl, and src oncogenes. J. Exp. Med. 164, 443-457. 19. Huitner, L, Szots, H., Welle, M., Van Snick. J., Moeiler, J. and Dormer, P. (1989) Mouse bone marrow-derived interleukin 3-dependent mast cells and autonomous sublines produce interieukin 6. Immunology 67, 408-413. 20. Kemp, D.J.. Harris, AW. and Adams. J.M. (1980) Transcripts of the immunoglobulin 04 gene vary in structure and Splicing during lymphoid development. Proc. Natl. Acad. Sci. USA 77, 7400-7404. 21. Klinken, S.P., Alexander, W.S. and Adams, J.M. (1988) Hemapoletlc lineage switch: v—raf oncogene converts 51-myc transgenic B cells into macrophages. Cell 53, 857-867. 22. Marrack, P., McCormack, J. and Kappler, J. (1989) Presentation of antigen, foreign major histocompatibility complex proteins and self by thymus cortical epithelium. Nature 338, 503-505. 23. McCulloch, EA. (1983) Stem cells in normal and leukemic hemopolesls. Blood 62, 1-13. 24. Mosmann, T. (1983) Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J. Immunol. Meth. 65. 55-63. 25. Parnes, J.R., Velan, B., Felsenfeld, A., Ramanathan, L, Ferrini, U., Appella, E. and Seidman, J.G. (1981) Mouse Bz-mlcroglobulln cDNA clones: a screening procedure for cDNA clones corresponding to rare mRNAs. Proc. Natl. Acad. Sci. USA 78, 2253—2257. 26. Ralph, S.J., Thomas, M.L., Morton, CC. and Trowbrldge, LS. (1987) Stmctural variants of human T200 glycoprotein (leukocyte common antigen). EMBO J. 6, 1251-1257. 27. Raschke, W.G., Baird, 8., Ralph. P. and Nakoinz, l. (1978) Functional macrophage cell lines transformed by Abelson leukemia virus. Cell 15, 261-267. 28. Rave. N., Ckvenjakou. R. and Blodtker. H. (1979) Identification of procollagen mRNAs transferred to DBM paper from formaldehyde agarose gels. Nucl. Acids Res. 6, 3559-3567. 29. Rigby, P.W., Dieckmann, M., Rhodes, C. and Berg, P. (1979) Labeling deoxyrlbonuclelc acid to high specific activity in vitro by nick translation with DNA polymerase I. J. Mol. Biol. 113, 237-251. 30. Rogers, J., Early, P., Carter, C., Calame, K, Bond, M., Hood, L and Wall, R. (1980) Two mRNAs with different 3’ ends encode membrane-bound and secreted forms of immunoglobulin a chain. Cell 20, 303-312. 31. Saga, Y., Tung, J.S., Shen, F.W. and Boyse, EA. (1987) Altematlve use of 5’ exons In the specification of Ly—5 isoforms distinguishing hematopoietic cell lineages. Proc. Natl. Acad. Sci. USA 84. 5364-5368. 144 32. Schwartz, R.G., Sonenshein, G.E., Bothweli, A and Gefter, ML (1981) Multiple expression of lg k-chain encoding RNA species in murlne plasmacytoma cells. J. Immunol. 126, 2104-2108. 33. Schwartz, R.C., Stanton, L.W., Riley, S.G., Marcu, KB. and Witte, O.N. (1986a) Synergism of v-myc and v-Ha-ras in the in vitro neoplastic progression of murlne lymphoid cells. Mol. Cell. Biol. 6. 3221—3231. 34. Schwartz, R.G., Stanton, L.W., Marcu, KB. and Witte. O.N. (1986b) An In vino model for tumor progression in murlne lymphoid cells. Curr. Top. Microbiol. Immunol. 132, 75-80. 35. Seidman, J.G. and Leder, P. (1978) The arrangement and rearrangement of antibody genes. Nature 276, 790-795. 36. Sheng-Ong, G.LC., Holmes. KL and Morse, H.C. (1987) Phorbol ester-induced growth arrest of murlne myelomonocytic leukemic cells with virus disrupted myb locus is not accompanied by decreased myc and myb expression. Proc. Natl. Acad. Sci. USA 84, 199—203. 37. Silver, J., and Kozak, C. (1986) Common proviral integration region on mouse chromosome 7 In lymphomas and myelogenous leukemias induced by Friend murlne leukemia virus. J. Virol. 57. 526-533. 38. Southern, EM. (1975) Detection of specific sequences among DNA fragments separated by gel electrophoresis. J. Mol. Biol. 98, 502—517. 39. Springer, T., Galfre, G., Secher, 0.8. and Milsteln, C. (1979) Mac-1: a macrophage differentiation antigen identified by a monoclonal antibody. Eur. J. Immunol. 9, 301-306. 40. Stanton, LW., Fahrfander, P.D., Tesser, PM. and Marcu, KB. (1984) Nucleotide sequence comparison of normal and translocated murine c-myc genes. Nature 310, 423-425. 41. Streuli, M., Hall, L.R.. Saga, Y., Schlossman, SF. and Salto, H. (1987) Differential usage of three exons generates at least five different mRNAs encoding human leukocyte common antigens. J. Exp. Med. 166, 1548-1566. 42. Thomas, M.L, Reynolds, P.J., Chain, A., Ben-Neriah, Y., and Trowbrldge, LS. (1987) B-cell variant of mouse T200 (Ly-5): evidence for altematlve mRNA splicing. Proc. Natl. Acad. Sci. USA 84. 5360-5363. 43. Thomas, P. (1980) Hybridization of denatured RNA and small DNA fragments transferred to nitrocellulose. Proc. Natl. Acad. Sci. USA 77, 5201-5205. 44. Walker, E., Warner, N.L., Chesnut, R., Kappler, J. and Marrack, P. (1982) Antigen-specific, I region-restricted interactions in vitro between tumor cell lines and T cell hybridomas. J. Immunol. 128, 2164-2169. 45. White, J., Hasklns, KM., Marrack, P. and Kappler, J. (1983) Use of l region-restricted, antigen-specific T cell hybridomas to produce idlotyplcally specific anti-receptor antibodies. J. lmmunol. 130,1033-1037. 46. Whitlock, C.A., Ziegler, S.F., Treiman, LJ., Stafford, J.l. and Witte, ON. (1983) Differentiation of cloned populations of immature B cells after transformation with Abelson murlne leukemia virus. Cell 32, 903-911. 47. Yam, L.T., Li, CY. and Crosby. W.H. (1971) Cytochemical identification of monocytes and granulocytes. Am. J. Clin. Path. 55, 283-290 APPENDIX 8 Introduction: This appendix presents “data not shown“ pertinent to the manuscript that comprises Chapter 2. These include data that demonstrated (i) that the tumors derived from R2 possess a more transformed phenotype than the R2 cell line; 0i) that alternative mechanisms of c-myc gene activation may occur in tumors derived from R2; (iii) the capability of the pre-B cells to undergo further differentiation during the process of tumorigenesis. A list of probes of oncogenes, tumor suppressor genes, growth factor genes, and sites of frequent viral integration used to screen for additional genetic lesions in the tumor cells is also included (Table 2). Materials and Methods: Cell culture. Cells were cultured under conditions described in Chen et al. (1991)(Chapter 2). Recombinant IL—7, a generous gift of Dr. Steven Gillis (lmmunex Corp., Seattle), was used in the culture of cells in the absence of feeder layers. Tumor challenges. Cells were washed twice in RPMI 1640 and were then resuspended in the same at 8x106 cells per ml. BALB/c mice, 4 weeks old, were injected intraperitoneally with either 0.25 ml or 0.5 ml of the cellular suspension. Animals were observed for a maximum of 10 weeks postinjection. Animals were sacrificed and autopsied when they become moribund or at 10 weeks. Latency in these experiments refers to the time until the animals became moribund. 145 146 Nucleic acid analyses. All hybridization analyses were performed as described in Chen et al. (1991)(Chapter 2). The kappa probe is a 3.4 kb Hindill restriction fragment derived from a plasmid (pcK), which contains a genomic fragment corresponding to kappa constant region sequences (Seidman and Cedar, 1978). Nuclear run-on experiments. Nuclear run-on experiments were performed essentially as described in Stewart et al. (1987) with the following modifications. Labeling was accomplished through the incorporation of (a -32P)UTP (600 Ci/mmol; Amersham). Isolated RNA was partially hydrolyzed in NaOH in order to allow differential analysis of initiation and elongation of c—myc transcription. RNA was resuspended in 250 pl of 20 mM HEPES (pH 7.5), 5 mM EDTA. 62.5 ul of 1M sodium hydroxide was added and the mixture was incubated on ice for 10 minutes. The reaction was quenched with 125 pl of 1M HEPES (free acid; pH 5.5). Hybridizations were carried out in 50% formamide, 5 x SSC, 5x Denhardt’s solution, 0.1% SDS. 50 mM sodium phosphate (pH 6.8), 250149/ ml single-stranded salmon sperm DNA, 5% dextran sulfate for at least 40 hours at 42° C. Hybridizations were washed to a stringency of 0.1 x SSC at 55° C. Single-stranded M13 probes were a generous gift of Dr. Alain Nepveu (Ludwig Institute for Cancer Research, Montreal). The exon 1 c-myc probe contained the 0.5 kb BamHl-Bglll fragment of the murine crmyc gene. The exon 2 and 3 probe contain a 3.4 kb BamHl-Hindlll fragment of the murine crmyc gene. The antisense control contained the 145 bp HaellI-Hindlll fragment at the 5’ end of the murine c—myc gene. The rGAPDH probe contained the 1.3 kb cDNA of rGAPDH. mRNA turnover assay. R2 cells and tumor cell lines were cultured as described in Chen et al. (1991)(Chapter 2), and maintained in an actively growing state by 147 addition of fresh media 24 hours prior to harvest. Cells were harvested, and counted with trypan blue staining to assess their viability. 1.5 x 10" cells were resuspended in 60 ml standard medium supplemented with Actinomycin D at a concentration of 50 micrograms per ml, and then split into three portions. The first 20 ml were immediately subjected to cytoplasmic RNA preparation by methods described in Chen et al. (1991)(Chapter 2), while the other two portions were incubated for either 1 /2 hour or 1 1 /2 hours after the addition of Actinomycin D. RNA of R2 and the tumor cell lines isolated at each time point was analyzed by a Northern blotting procedure as described in Chen et al. (1991)(Chapter 2). Northern blots were sequentially hybridized to probes for c-myc and rGAPDH. The stability of c-myc RNA in R2 and the tumor cell lines was assessed by their relative decrease in abundance over time by the method described in the table legend. Results: The tumors possess a more transformed phenotype than their parental cell line. As described in the Chapter 2, the infrequent occurrence and latency of tumors derived from the R2 cell line raised the question of whether tumor progression required the acquisition of genetic lesions in addition to v-Ha-ras expression. Other explanations for the weak tumorigenicity of the R2 cell line are plausible. First, it is possible that R2 cells are normally able to grow into tumors if they do not acquire other genetic lesions, which may somehow stimulate the host immune system to eliminate or inhibit their growth. This scenario is unlikely, since Schwartz et al. (1986b) found R2 to be weakly tumorigenic in athymic nude 148 mice. Second, the infrequent occurrence may relate to the variation of epigenetic factors among hosts, and the long latency may only reflect the in vivo slow growth properties of R2 cells. In order to ascertain whether the tumor cells had gained growth properties consistent with tumor progression, the growth properties of early passage tumor cell lines were compared to the R2 parental line. Two growth properties were tested: the ability to grow in vitro independent of adherent feeder cells and the ability to generate tumors in syngeneic mice. R2 had previously been found to be dependent on an adherent cellular feeder layer for growth (Schwartz et al., 1986a). All of the tumor cell lines grew well on feeder layers. In order to test whether the tumors had gained some level of growth factor independence, the tumor cell lines and R2 were cultured in the absence of feeder layers. While R2 was incapable of growth in the absence of a feeder layer, all of the tumor cell lines exhibited growth (Figure 1). This is most consistently the case at higher cell density. Since lL-7 has been reported to be required for the growth of pre-B cells (Namen et al., 1988), it was of interest to test the lL-7 responsiveness of R2 and the tumor cell lines derived from it. R2 (Figure 1) and other v-Ha-ras-transformed pre-B cell lines (data not shown) are both dependent on and dramatically responsive to lL-7 for growth in the absence of a feeder layer. Several of the tumor cell lines, although independent of cellular feeder layers and lL-7 for growth, retained some responsiveness to lL-7 (Figure 1). The abilities of the tumor cell lines to form tumors in syngeneic BALB/c mice were compared to that of the parental R2 cell line. Tumor cell lines 1, 2, 3, 4 and 149 /f (ILLS -./////‘ .”*.._._._. "— ‘03 M ‘4 5.1 . *m-U *omm OIL? Figure 1. Growth of the R2 and seven tumor cell lines (1-7) in the presence and absence of lL-7. Cells were removed from feeder layers and resuspended in fresh medium with and without 10 units/ml IL-7. Either 5x10a or 5x104 cells were plated in suspension in 1 ml. On day 4, viable cells were counted in the presence of trypan blue. The graph plots the average total cells of duplicate cultures. 150 Table 1. Tumor challenges in BALB/c mice. Cell Line Frequency (animals) Average Latency Till Moribund (days) 2x106 cells“ 4x106 cells‘ 2x106 cells‘ 4x106 cells“ R2 1 /4 1 /4 81 70 1 1 /5 5/5 44 25 2 2/5 5/5 41 24 3 1 /4 5/5 44 27 4 4/4 ND. 22 ND. 5 ND. 3/5 ND. 50 6 ND. 1 /5 ND. 22 7 2/2 3/4 16 45 N.D. = Not determined ‘Two inocula were used. 7 were dramatically more tumorigenic than R2 (Table 1). Tumor cell lines 5 and 6 had lower frequencies of tumor induction than the other tumor cell lines. This is consistent with their lack of growth in the absence of a feeder layer in experiments of longer duration than those presented in Figure 1 (data not shown). All of the mice challenged with tumor cell lines did have a shorter latency till moribund than did those challenged with R2 (T able 1). The data on feeder layer independence and tumorigenicity demonstrate that the tumor cell lines have acquired transformed growth potential which exceeds that of their parental cell line. To examine the molecular events that underlie the transformed phenotypes of tumor cells, we have used a set of probes of oncogenes, tumor suppressor genes, growth factor genes, and sites of frequent viral integration (Table 2) to test for their involvement. Gene rearrangement or altered expression of these genes or sites 151 .co_mmocaxo .anx..nx ..uom..om ....nc.=.n= .9505? .3 38.35.39. ¢ouu.>¢ Loan» >.cou ..¢oum ..¢ b 636: o... .8533 9.0... m..3uo> «5 so...) 5.... 2.3.8 .3 «35.39. 2.53.5.0 .303 505.8: co 6339.98 .933 <5. .8 5232.3 0.538.023.53. «33.3., 5...: 3o... 53. ucueoucoccao. .9533 ..o 303 50533 :0 tenuous“. .n o.nn< .n Lao:u_.04 caufip...o uo mxmcounx moo..... .3 5523.6 .1 .3395 32.... so 2695.. .o.z 6:6.6666 .o.z uca.oaou .32.... 63:53. 38...... .3355 .a cuscom_m .3 cgdommw .: .65.. .: o.o..oas. .x 0....485. .66 amaz< .o as..: .x 6.66 gene. xcoLL uu.< Loncmm ..s.< .u Lump—m .:.¢ Lacuna..o. .x.¢ Luuugtoa .o cass.m .o ouu_3 OUSLOwflu .a.h 0 6.9.3 mm .8 c3833.... .855 of E 3302.. 3 >2. 35 355 o_u:n .636 6625 «came -n¢o¢ opm.q can -n¢oa -nama .4..~Lxmua umsma.c moo n_aumo:.mn oc>moua oc>moua n.u:n Nwmama magma -n¢ma n—uaa npuan mmnmmq m.~.nau:a .ouuo> n=-_umm ax n.o .uma.n= ax o.« .c ax ~.o _¢._umm ax c.— .xm ox no.o _¢-m: ax o." n: ax m.n n: nx o.~ __:>¢-_¢ ax N.— .¢ 6. mm.o .3 a. o.n .a a. 4.~ .:¢ 6. L.~ .g a. 5.8 .ocx ax ~.n .36. a. m.o .um a. m.o .oUz-_uma nx 0.9 __a>< as mm.o .u ox o.~ .m nx e.~ _¢ ax m.o _:m-_u.u nx N.N .a 93 n.p .m 93 ~.p __:>a nx 0.9 _¢._nx ax o.~ >¢..=a .a .xu..cax .cax >¢..¢ .cax .>¢..=m..¢ AucoEmacevonocm 33:33 «>333 05 Lo» 9.29.9.5 E .zu .um..=a .zo .¢ .zm..a <3... .5... <2“ .a..=m .s..=m .c..=m ¢ .u..=m .a..=m .zu oe>~c n m m.~ c.s o.o~A o.o~ o.- o—A oémA «.m— m.P~ o.o~A o.o~w o.o~A c.opx c.o~A o.o~A o.—P o.oa c.- bo.o~A no.o~a o.¢~A o.c—A o.-A c.o~A o.opx o.-A “savoucom p-_;a —._>o P-u>n ~.E_q —-e_o ¢-_>.e n-_>.e ~-_>.e —-_>.E xae ommm u< .mmo.uu=x nmo ax u_x h-.— —-mmu m-.— mmuzu woc-u n>E-u u>E.z msw-> xu. xuc o.m.> .no.> acme vow: 9.0: =5 «39:. .~ 033 152 was examined by Southern and Northern blotting analyses. Tumor 4 showed altered expression of several genes, and was described in detail in Bretz et al. (1992)(Appendix A). The c-myc gene was aberrantly expressed in four of the seven tumor cell lines tested (two of these also showing retroviral integration at c- myc) as described in Chen et al. (1991)(Chapter 2). No gene rearrangement was found in about a 20 kb region surrounding most of the other oncogenes or frequent viral integration sites tested in the tumor cells. No altered expression of other tested genes was detected in the tumor cells, with the exception of Tumor 4 (see Chapter 4). Studies of mechanisms of elevated c-myc expression We have observed a 3-fold increased level of c-myc mRNA in tumors 1 and 3, presumably as a result of the proviral insertion, and 4-fold elevated levels of c-myc mRNA in tumors 6 and 7 in the absence of any obvious genetic alteration (Figure 6 in Chapter 2). The c-myc mRNAs were all approximately 2.4kb in length, suggesting normal promoter usage and an unaltered RNA structure (Figure 6 in Chapter 2). RNAse protection studies confirmed normal promoter usage (data not shown). Nuclear run-on experiments were performed in order to examine (i) whether the elevated c-myc mRNA levels reflected increased transcription and (Ii) the role of transcriptional attenuation (Bentley and Groudine, 1986) in modulating c-myc expression in these tumors. Transcriptional initiation of c-myc was evaluated by hybridization of labeled RNA to a probe for exon 1 of c-myc, while elongation of transcription was evaluated by hybridization to a probe for exons 2 and 3 of c-myc. 153 R2 MYC EXON I MYC EXON 2&3 ANTISENSE rGAPDH TRANSCRIPTION LEVELS MYC EXONI 1.0 3.1 3.1 1.6 1.0 MYC EXON 28:3 1.0 2.0 1.5 2.0 1.2 Figure 2. Nuclear run-on transcription. Labeled nuclear run-on products from nz and tumors 1, 3, 6 and 7 were hybridized to probes for c-myc exon 1, c-myc exons 2 and 3, antisense upstream of c-myc and rGAPDH. Relative levels of transcription were determined by densitometry and normalized to rGAPDH. 154 Densitometric values were normalized to the amount of hybridization detected to a probe for rGAPDH. Background hybridization was evaluated with a probe for anti-sense transcripts upstream of the c-myc locus. In comparison to R2, tumors 1 and 3 showed about 3-fold increased transcriptional initiation and about 1.5 to 2-fold increased elongation (Figure 2). Since these values average the activity of the retrovirally activated allele(s) with the normal c-myc allele, they are likely to underestimate the degree to which the presence of the MoMuLV LTR affects c-myc transcription. The elevated transcription detected in this experiment is consistent with the 2 to 3-fold elevated steady state levels of c-myc mRNA observed by Northern analysis (Figure 6 in Chapter 2). Since initiation of transcription increases more than elongation, increased initiation rather than decreased attenuation is the probable mechanism by which retroviral integration increases c-myc mRNA expression. Tumor 6 showed about 1 .5-fold increased initiation and about 2-fold increased elongation (Figure 2). This is consistent with the 3-fold increased steady state levels of c—myc mRNA observed for tumor 6 in Figure 6(of Chapter 2). On the other hand, tumor 7 did not show significant increases in either parameter. The data offer no evidence for a transcriptional mechanism leading to the elevated c-myc mRNA levels observed for tumor 7 (Figure 6 in Chapter 2). Determinations of the half-life of c-myc mRNA in the presence of actinomycin D did not reveal significant differences between R2 and tumors 1, 3, 6, and 7 (Table 3). R2 and tumor cell lines were expanded and maintained in an actively growing state, and then treated with 5 microgram/ml Actinomycin D. Portions of cells were then harvested at three time points: 0 hour, 1 /2 hour, and 1 1/2 hour after adding Actinomycin D. RNAs of each cell line were isolated at the 155 three time points and subjected to Northern hybridization analyses with a c-myc- specific probe and a rGAPDH gene probe. Densitometric measurements were taken for both c-myc and rGAPDH specific signals on the resulting autoradiogram. Since the level of rGAPDH remained relatively unchanged among R2 and tumor cell lines during the 1 1 /2 hour time period, we normalized the level of c-myc mRNA in each time point to that of rGAPDH at the same time point under the assumption that the degradation rate of rGAPDH mRNA is the same in R2 and tumor cell lines. The stability of c-myc mRNA was determined by the assessment of the relative half-life assigned to R2 and each tumor cell line by the method described in the table legend. The relative T 1 /2 of c-myc mRNA is 32 minutes, whereas those of tumor cell lines ranged from 28 to 40 minutes. Therefore, we concluded that alterations in the stability of c-myc mRNA did not play a role in the increased c-myc mRNA levels observed in these tumor cells. Table 3. Relative stability of c-myc mRNA in R2 and tumors cells relative T 1 /2 (min)* n2 32(29,34) T1 40(35,4e) T3 36(31,42) T6 28(26,29) T7 38(35,41) *These values were determined by the following calculation procedure. The signal of c-myc was normalized to rGAPDH for a loading control, and then the resulting values were plotted on a semilog graph (log of hybridization intensity versus time (minutes)). The relative T 1 /2 was determined by the time required for the loss of half of the original hybridization intensity. 156 Observatlon of light chain rearrangement In tumor cell llnes One other interesting observation obtained in these studies was the detection of immunoglobulin light chain rearrangements in most of the tumors (Figure 3). As mentioned in Chapter 2, all tumor cell lines possess the same immunoglobulin (lg) heavy chain rearrangements as that of the parental R2 cell line (Figure 4). A southern blot hybridization analysis with a probe specific to the J region of lg heavy chain revealed hybridizations to a 6.0 kb germ line fragment (Figure 4, lane 1) on EcoRl digested liver DNA and to one or two rearranged fragments (7.0 kb and 3.5 kb, Figure 4, lane 2-9) on DNA of the R2 and tumor cells. DNA from the original tumor sample (a portion of which may consist normal cells) of tumor 6 was used in the analysis, therefore, a germ line fragment was also observed (lane 8). When the same DNAs were digested with BamHI, and probed with a DNA fragment from the constant region of the kappa chain, all the tumor cell lines showed dramatically different banding patterns (Fig. 3, lanes 2, 4, 5, 7, and 8) from the germ line fragment observed in the R2 cell line and liver (Fig. 3, lanes 1 and 9, respectively). Tumor cell lines 2 and 5 (Fig. 3, lanes 3 and 6, respectively) both retained the germ line fragment, but a portion of these cells may already have undergone kappa chain rearrangements as judged by the presence of some minor hybridized bands. No lambda light chain gene rearrangement was found in the R2 and tumor cell lines (data not shown). Immunopreoipitation experiments with both anti-mu and anti-kappa chain anti-sera revealed that the rearrangements successfully produced authentic immunoglobulin heavy and light chain proteins(data not shown). 157 KAPPA 32.1.2fi34 56 7L 1 ;: ."'-' 235 j 5; c 9.4 >, . 6.6 b, In» 4.4 ,- 2.0> Figure 3. Kappa chain gene rearrangement. BamHl digested R2 (lane 1), liver (lane 9), and tumor cell DNAs (lane 2-8) were subjected to gel electrophoresis through 0.8 % agarose, blotted to nytran paper, and hybridized with a nick- translated probe of a kappa-chain-specific DNA fragment. Size markers are the positions of an ethidium bromide-stained Hindlll digest of bacteriophage lambda and are denoted in kilobases. 158 1 2 3 4 5 6 7 8 9 23>!” * A. 9.4» 65> v " T "" u '41» 23> 2.0V Figure 4. Mu chain gene rearrangement. EcoRl digested liver (lane 1), R2 (lane 2), and tumor DNAs (lane 3-9) were electrophoresed through a 0.8 % agarose gel. The blot was probed for the J region of mu heavy chain gene. Size markers are the positions of an ethidium bromide- stained Hindlll digest of bacteriophage lambda and are denoted in kilobases. 159 Discussion: The data presented here confirm that the R2 tumors had acquired a more transformed phenotype as evidenced by their high tumorigenicity and their growth factor independence. The fact that these tumors were not uniformly tumorigenic (i.e. did not cause tumors in all the tumor-challenged mice) suggested the possibility of tumor regression through a variety of mechanisms. These may include stimulation of host immune system perhaps by generating a recognizable tumor specific antigen, or growth inhibition by some intrinsic factors that evolved in tumor cells during the course of tumor challenge. The intermediate transformed phenotypes of these tumors may provide tools for studying tumor progression. The regulatory mechanisms responsible for increased levels of c-myc mRNA probably occur at both transcriptional and posttranscriptional levels as described by Klein and Klein (1985). In the case of tumors 1 and 3, MoMuLV had integrated immediately 5’ to c-myc in a reverse transcriptional orientation, suggesting enhancer activation. Nuclear run-on experiments showed that these tumors with MoMuLV integration near c-myc displayed increased levels of transcription consistent with the increased steady state level of c-myc mRNA. The nature of the events leading to elevated levels of c-myc mRNA in the other two tumors is unclear. Tumor 6 displayed increased transcription, but no gross structural alterations were observed on either the DNA or RNA levels. The attenuation of transcription in the first exon of c-myc was also unaffected. Tumor 7 showed no increase in transcription and, as in the case of tumor 6, there were no structural abnormalities observed in either the c-myo gene or its transcription products. Southern blot analyses of genomic restriction fragments extending approximately 160 15 kb upstream and 30 kb downstream of c-myc detected no rearrangements in either tumor 6 or 7 (data not shown) suggesting that retroviral integration is unlikely to play a direct role in the activation of c-myc in these tumors. On the other hand, MoMuLV integrations at MIvi-1 and MIvi—4 can affect c-myc over a long distance (Lazo et al., 1990). However, we have not detected rearrangements at these sites (data not shown). The increased c-myc mRNA level in tumor 7 may be due to posttranscriptional mechanisms such as increased maturation rate of poly A“ RNA and increased transport rate through the nuclear membrane. Studies of mRNA half-life show that these mechanisms are not involved. The mechanism by which c-myc activation promotes tumorigenesis is unclear. Expression of v-myc has been reported to abrogate the dependence of lymphoid and myeloid cell lines on lL-2 and lL-3 for growth (Payne et al., 1982). Since all the tumors with and without c—myc activation show reduced dependence on lL-7, it is difficult to conclude that a similar causal relationship exists between c-myc activation and alleviation of IL—7 dependence. The fact that pre-B cells were able to continue to undergo light chain rearrangements and to successfully produce kappa chains suggests that introduction of the v-Ha-ras oncogene does not block B cell differentiation. It also implies that B cell lymphomas may result from a gradual acquisition of genetic lesions by a precursor B cell that can progress along the differentiation pathway. Similar results had been shown in Eli-myc transgenic mice, in which tumors of both pre-B and B cells were obtained (Adams et al., 1985; Harris et al., 1988). On the other hand, the majority of tumors induced by v-abl seem to be frozen at a pre-B stage (Sklar et al., 1974; Sklar et al.,1975; Weimann, 1976). Interestingly, 161 long term cultures (as long as the period of tumor challenge) of the R2 cell line did not show evidence of light chain rearrangement in vitro (data not shown). It seems that some in vivo factors may be important for light chain rearrangement in addition to a successful heavy chain rearrangement. In vitro triggering of light chain rearrangements of R2 cells may help to identify these factors, and provide more insights on B cell differentiation. Although no gross gene aberrations in genes other than c-myc were detected in the R2-derived tumor cells, we can not exclude their roles in the additive transformed phenotypes of these cells. Mutations of other types such as point mutation, small deletion, or viral integration in regions outside of the tested regions may have occurred. Further investigations will be required to elucidate the secondary events in the tumor progression of v-Ha-ras transformed pre-B cells. 162 References: Adams J.M., A.W. Harris, C.A. Pinkert, LM. Corcoran, W.S. Alexander, S. Cory, R.D. Palmiter, and R.L Brinster. (1985). The c-myc oncogene driven by immunoglobulin enhancers induce lymphold malignancy in transgenic mice. Nature 318, 533-538. Bentley D.L, and MA. Groudine. (1986). A block to elongation is largely responsible for decreased transcription of c-myc in differentiated HL-60. Nature 321, 702-704. Bretz J. S.-C. Chen, D. Redenius, H.-L Chang, W.J. Esselman, and RC. Schwartz (1992). Uneage switch macrophages can present antigen. Develop. lmmunol. in press. Chen S.-C., D. Redenius, and RC. Schwartz. (1991). Tumorigenesis of a v-Ha-ras-expresslng pre-B cell line selects for c-myc activation. Biochem. Biophy. Res. Comm. 178, 1343-1350. Cuypers H.T., G. Selten, W. Quint, M. Zijlstra, E.R. Maandag, W. Boelens, P. van Wezenbeek, C. Mellef, and A. Bems. (1984). Murine leukemia virus-induced T cell lymphomagenesis: integration of proviruses in a distinct chromosomal reglon. Cell 37, 141-150. Graham M., J.M. Adams, and 8. Cory. (1985). Murine T lymphomas wlth retroviral inserts In the chromosomal 15 locus for plasmacytoma variant translocations. Nature 314, 740-743. Lazo P.A. , J.S. Lee, and RN. Tschilis. (1990). Long-distance activation of the myc protooncogene by provirus insertion in MIvi-1 or MIvi-4 in rat T-cell lymphoma. Proc. Natl. Acad. Sci. USA 87, 170-173. Mucenski M.L., B.A. Taylor, J.M. lhle, J.N. Hartley, H.C. Morse, Ill, N.A. Jenkins, and N.G. Copeland. (1988a). Identification of a common ecotropic viral integration site evi-1 in the DNA of AKXD murlne myeloid tumors. Mol. Cell. Biol. 8, 301-308. Namen A.E., A.E. Schmierer, C.J. March, R.W. Overell, LS. Park, D.L Urdal, and D.Y. Mochizukl. (1988). B cell precursor growth-promoting actlvlty: purification and characterization of a growth factor active on lymphocyte precursors. J. Exp. Med. 107, 988-1002. Payne G.S., J.M. Adams, and HE. Varmus. (1982). Multiple arrangements of viral DNA and an activated host oncogene in bursal lymphomas. Nature 295, 209-213. Schwartz R.C., LW. Stanton, S.C. Riley, KB. Marcu, and ON. Witte. (1986a). Synergism of v-myc and v-Ha-ras in the In vitro neoplastic progresslon of murlne lymphoid cells. Mol. Cell. Biol. 6, 3221-3231. Schwartz R.C., LW. Santon, S.C. Riley, KB. Marcu, and ON. Witte. (1986b). An in vitro model for tumor progression in murlne lymphoid cells. Curr. Topics Micro. Immunol. 132. 75-80. Sklar M.D., B.J. White, and WP. Rowe. (1974). Initiation of oncogenic transformation of mouse lymphocytes in vitro by Abelson Leukemia Virus. Proc. Natl. Acad. Sci. USA 71, 4077-4081. Stewart C.J., M. Ito, and SE. Conrad. (1987). Evidence for transcriptional and post-transcriptional control of the cellular thymidine kinase gene. Mol. Cell. Biol. 7, 1156-1163. Tsichlis P N., G. Strauss. and EH. Liu. (19833). A common region for proviral DNA integration in MoMuLV-induced rat thymic lymphomas. Nature 302, 445-449. Tsichlis P.N., P.G. Strauss, and CA. Kozak. (1984). Cellular DNA region involved in induction of thymic lymphoma(MIvi-Z) maps to chromosome 15. Mol. Cell. Biol. 4, 997-1000. Summary and Discussion The goals of my thesis study were to search for oncogenes that were capable of cooperating with the v-Ha-ras oncogene in transforming murine B cells, and to examine the differentiation status of tumor cells. Two model systems were used. The first model system tested for genes that could cooperate v-Ha-ras in transforming murine B cells in vivo, whereas the second model system examined this issue in vitro. In the first model system, we found that tumors had acquired more malignant growth properties by comparing the growth characteristics of R2, a v-Hr- ras-expressing pre-B cell line, and tumor cells derived from it. The tumor cell lines exhibited greater tumorigenicity and shorter latency in tumor development than the parental R2 cell line. In addition, tumor cell lines had acquired growth factor independence. Two major molecular events have been shown to be associated with these transformed phenotypes: an incgased number of virus-associated restriction fragments on southern blot analysis, and alteration of c-myc expression. First, studies on the viral integration patterns of R2 cells and tumors suggested that 6/7 tumors were an outgrth of a single R2 subclone, which contained unique viral related fragments. These unique virus-associated fragments could have resulted either from recombination of the original integration sites of the parental cell line, or from new viral integrations, or both. Oncogene activations had been found previously through each mechanism. We found two tumors that had activated c-myc by viral integration. Many, if not all, proviruses near c-myc have sustained deletions of the viral genome, probably as a result of homologous recombination between the two 163 164 two LTRs. Sometimes little more than a solitary LTR is left intact (Robinson and Gagnon, 1986). A mechanistic explanation of the requirement for these deletions was given by Cullen et al. (1984), who showed that transcription starting in the 3’ LTR of an intact provirus is quenched by transcription driven by the 5’ LTR. By artificial termination of transcription within the provirus, or by the deletions as found in tumors, the activity of the 3' LTR becomes sufficient to act as a strong promoter (Fujisawa et al., 1985) and thus to activate oncogenes. The c-myc rearrangement in two of the R2 tumor cell lines resembles the above observation in that the genome of MoMuLV integrated in the c-myc gene in these two tumors has undergone a further rearrangement (data not shown). This viral genome rearrangement was assessed by a similar approach to that described in Figure 4 (Chen et al., 1991), with the exception that restriction mapping utilized enzymes that cut inside the viral genome rather than in the LTRs. However, we do not know the significance of this viral genome rearrangement, since the activation of c-myc gene in this case is presumably through an enhancer activation rather than a promoter activation. The reintegration of MoMuLV and avian leukosis virus (ALV) into the cellular genome of infected cells has been well documented in the tumors they generate, and are thought to be causative for tumor formation through the activation of nearby cellular oncogenes (Jaenisch, 1976; Payne et al., 1982). Studies on transgenic mice carrying a single copy of MoMuLV DNA sequences has shown an increase to two MoMuLV specific DNA copies per haploid mouse genome in preleukemic tissues, and a further increase to 3-4 copies in leukemic tissues (Jaenisch, 1979). This amplification of MoMuLV is seen in target organs but not 165 in nontarget organs, and thus appears to be related to leukemic transformation. At least one tumor specific virus-related fragment (bone-1) in our studies belonged to the aforementioned class (reintegration), since DNA of the parental R2 cell line showed a germ line pattern at this locus. The nature of other tumor specific virus-related fragments is unknown. We were unable to detect any transcriptional activity over a 20 kb region in the vicinity of the bonc-1 locus. However, sequence homology of bonc-1 was found to genomes of many mammalian species and chicken. The high conservation of bone-1 suggests that this region has an important function. On the other hand, activation of protooncogenes through distal provirus integrations (270 kb) has been observed (Lazo et al., 1990), and it is possible that a putative oncogene is located beyond the 20 kb region studied. Whether this putative gene would indeed be able to cooperate with v-Ha-ras gene in course of tumor progression, and whether it would have B cell specificity in the induction of neoplasia remains to be tested. Although I was not able to definitively identify a novel oncogene during the period of my thesis study, the fact that MoMuLV was highly mobilized in infected cells provides a method to identify new protooncogenes involved in hematopoietic diseases. In retrospect, modifications of the insertional mutagen (MoMuLV) might have been added to facilitate the identification of viral integration sites. These modifications might include the addition of a bacterial supF gene or some other bacterial indicator gene to the MoMuLV sequences. Tagging with a supF gene would allow the construction of an integration library in a amber mutant 1. phage that could contain only clones with sequences of supF integrated proviruses and their flanking cellular loci as described by Shih et al. (1988). 166 The second molecular event found among the tumor cell lines derived from the R2 cell line was the increased steady state level of c-myc mRNA in four of the seven tumors. Studies of c-myc activation have revealed that the c-myc gene can be deregulated by several mechanisms: increased transcription, increased elongation, and perhaps increased transport through nuclear membranes (for review see Klein and Klein, 1985). Our findings are consistent with varied mechanisms for deregulation of c-myc. Two of the four tumors studied here had suffered a MoMuLV integration at about 150 bp 5’ of the first exon of the c-myo gene in a reverse transcriptional orientation, presumably driving the expression of the c-myc gene through enhancer activation and increased transcription. The exact mechanism of c-myc activation of the other two tumors is unknown, although one tumor appears to be transcriptionally activated. Our finding of MoMuLV integration into the c-myc gene in B cell lymphomas is novel. MoMuLV integration in the c-myc locus has been commonly found in murine thymomas (Selten et al., 1984). Feline leukemia virus integration near c- myc has been observed to occur only in T cell lymphoma (Neil et al., 1984). Mucenski et al. (1987) reported that viral integration in the c-myc locus occurs only in murine T cell lymphomas from studies on a panel of B cell and T cell lymphomas from AKRD mice. Our finding suggests that the tissue tropism of MoMuLV (and perhaps other MLVs) in naturally occurring tumors O.e., thymomas) may not be attributable to tissue specific integration sites. The observation that viral integrations into pim-1, originally identified as a common T cell integration site, were also detected in pre-B, and B cell lymphomas supports this idea (Mucenski et al., 1987). Although some virus integration sites such as mlvi-1, mlvi-2, fis-1, 167 and pvt-1 have been shown only found in T cell lymphomas (Mucenski et al.1987), the significance of these loci in tumorigenesis is not clear since no gene products have been found. C-myc gene alterations have been associated with many B cell neoplasias and have been proven to be able to initiate or promote B cell malignancy in transgenic mice (Adams and Cory, 1991). Cooperation of the c-myc gene with the v-Ha-ras gene in transformation has been shown in both fibroblasts (Land at al., 1983a) and B lymphoid cells (Schwartz et al., 1986a). The occurrence of c-myc alteration in tumor challenges with a v-Ha-ras-expressing pre-B cell suggests the validity of this model system in identifying genes that are associated with the in vivo tumor progression of B cell neoplasia. Besides retroviral integration and c-myc activation, we did not detect other gene rearrangements in tumors when probes of other known oncogenes, tumor suppressor genes, growth factor genes, and flanking genes of frequent viral integration sites (as listed in Appendix B) were used in southern hybridization analyses. Another approach applicable to the search for non-virally related secondary events involved in tumor progression of these tumors is the transfection of tumor DNA into indicator cells. If this approach were taken, care would have to be taken in choosing a appropriate indicator cell, since the tumor cells all posses a v-Ha-ras gene which is known to be dominant in focus formation assays in NlH3T3 cells. Two other interesting features were observed among the v-Ha-ras tumors: a continuation of B cell differentiation and a lineage switch to macrophage-like cells. As discussed in appendix B, most tumors derived from the pre-B cell line, 168 R2, had undergone kappa chain rearrangements and produced kappa chain proteins. However, we did not observe detectable light chain rearrangements in R2 cells that had been cultured in vitro for a similar or longer period of time as that required for tumorigenesis. This discrepancy was particularly interesting to us, since it is generally thought that light chain rearrangements will be triggered once a successful heavy chain rearrangement has been generated in the same cell. These results suggested that additional factors are required for light chain rearrangements besides the production of an authentic heavy chain protein. This factor was absent in the Whitlock-WItte culture system but present in mice. A similar lack of in vitro light chain rearrangement was also found in another v-Ha-ras transformed cell line, R1, which was generated by the same procedure as that of R2. Tumors derived from R1 cells also exhibited light chain rearrangement (data not shown), suggesting the generality of this phenomenon. On the other hand, it is also possible that some inhibitory factors may be present in the Whitlock-Witte culture system to prevent differentiation of these pre-B cell lines. A similar phenomenon has been demonstrated by Alt et al. (1981) on cell lines derived after in vitro infection of bone marrow or fetal liver cells with Abelson murlne leukemia virus (A-MuLV). Most, if not all, of the cell lines were pre- 8 cells and they rarely went through light chain rearrangement, even after a long period of cultivation in vitro. In contrast, tumors induced by A-MuLV often display light chain rearrangements (Sklar et al., 1975; Weimann, 1976). However, the fact that certain subclones of pre-B cell lines established in the Whitlock and Witte are capable of undergone light chain rearrangements in vitro (Denis and Wm, 1986) argues against this possibility. Perhaps, this in vitro inhibition of light chain 169 rearrangements in both v-Ha-ras and v-abl expressing pre-B cells are associated with these oncogene expression. An in vivo factor may bypass this inhibitory effect of oncogene expression in tumors carrying the same oncogenes. At any rate, these v-Ha-ras transformed cell lines provide a good model system to search for factors that may interfere or participate in the differentiation process of B cells. Another interesting feature of these studies was the discovery of a lineage switch of certain B cell tumor lines to macrophage-like cells. Although other groups have reported a similar phenomenon (Klinken et al., 1988; Davidson et al., 1988; Borzillo et al., 1990), the studies presented here uniquely establish the functionality of these lineage switch macrophages: LPS-induced cytokine release and the ability to present antigen. These macrophage cell lines may be the first macrophage lines ever reported to present antigens in vitro (John Cohen, U. Colorado, personal communication). Therefore, they are potentially useful in research areas such as of B cell activation, clonal expansion of T cells, and other cell-cell interactions that require macrophages with an antigen presenting capacity. They and their antecedent cells (the R1 and R2 cell lines) may prove useful in studying the lineage determination of lymphoid and myeloid cells. A second model system which involved the introduction of oncogenes into v-Ha-ras transformed cells or the co-infection of bone marrow cells with retroviruses carrying a v-Ha-ras oncogene and another oncogene of interest provided a convenient way to test the oncogenic potentials of various oncogenes. With this model, we have shown that expression of lL-7 was not sufficient to cause a fully tumorigenic phenotype in a v-Ha-ras expressing cell line. These findings 170 were consistent with what had been reported by Young et al. (1991) and Overell et al. (1991). Both groups reported that IL-7 can participate in the tumorigenesis of murine B cell neoplasia, with the requirement of other secondary events. The insufficiency of lL-7 expression to confer tumorigenicity may imply the existence of a tightly regulated mechanism of lL-7 induced growth in vivo. Another important finding from this model system was the discrepancy between the highly tumorigenic phenotype of cells transformed by a co-infection of bone marrow with v-Ha-ras and myc, or v-Ha-ras and lL-7 and the non- tumorigenic phenotype of cells sequentially infected with the same two oncogenes. The clonality of the outgrowing cell lines in the co-infection of bone- marrow and their high frequency and short latency for generation of tumors contrast sharply with the properties of v-Ha-ras cell lines superinfected with v-myc or lL-7 viruses. The clonal outgrowths of co-infections of bone marrow may represent the dominance of a rare and more highly transformed cell over many other cells carrying one or both oncogenes in this heterogeneous population. While lL-7 and v-Ha-ras expression may contribute to the transformed phenotype, the cooperation of other rare oncogenic events is critical to the tumorigenic phenotype. The more homogeneous population in the sequential infection of a single cell line does not as easily allow selection for other rare events. Therefore, interpretation of experiments of this sort should be made carefully. To ensure that the synergistic effect of two oncogenes in any particular system is indeed derived from the two oncogenes and not other "co-selected events“, the two oncogenes should be sequentially introduced into a cell line rather than a heterogeneous population. Cells with an intermediate transformed phenotype derived from the 171 these experiments can be used as targets for testing the involvement of other oncogenic events. in summary, results from both of our model systems for B cell neoplasia suggested that multiple events (at least three) were required for the induction of a fully malignant B cell phenotype. Both the c-myc gene and lL-7 gene were capable of cooperating with the v-Ha-ras oncogene in transforming murine B cells. However, to achieve a fully tumorigenic phenotype, a tertiary event(s) was required in both instances. The two models systems provide the opportunity to identify this tertiary event (3) as well as other independent steps involved in the tumor progression of B cell neoplasia. They also provide good tools to study B cell differentiation and lineage determination of myeloid and lymphoid cells. In the future, if particular gene alterations are found to be restricted to the development of murine B cell neoplasia, and a correlation is found between murine and human B cell neoplasia, detection of early-stage neoplastic cells may be possible. Detection protocols may operate through the identification of mutant (quantitatively or qualitatively) gene products secreted into the blood or other body fluids, or through the detection of antibodies to the mutant gene products. In addition, the presence of genetic alterations in tumors may provide a molecular tool for improved prognostic evaluation of patients, as is now possible with colorectal cancer (Vogelstein et al., 1989; Kern et al., 1989). Finally, the identification of mutant gene products in tumors may provide targets for new chemotherapeutic agents. "‘illllilllllllli