@ Oncogene (2000) 19, 1002-1009 © 2000 Macmillan Publishers Ltd All rights reserved 0950-9232/00 $15.00 www.nature.com/onc Use of MMTV-Wnt-1 transgenic mice for studying the genetic basis of breast cancer Yi Li*', Wendy P Hively' and Harold E Varmus' ‘Division of Basic Science, National Cancer Institute, 49 Convent Drive, Building 49, Room 4A56, Bethesda, Maryland, MD 20892, USA Wat-1 was first identified as a protooncogene activated by viral insertion in mouse mammary tumors. Transgenic expression of this gene using a mouse mammary tumor virus LTR enhancer causes extensive ductal hyperplasia early in life and mammary adenocarcinomas in approxi- mately 50% of the female transgenic (TG) mice by 6 months of age. Metastasis to the lung and proximal lymph nodes is rare at the time tumors are detected but frequent after the removal of the primary neoplasm. The potent mitogenic effect mediated by Wat-1 expression does not require estrogen stimulation; tumors form after an increased latency in estrogen receptor o-null mice. Several genetic lesions, including inactivation of p53 and over-expression of Fgf-3, collaborate with Wat-1 in leading to mammary tumors, but loss of Sky and inactivation of one allele of Rb do not affect the rate of tumor formation in Wat-1 TG mice. Oncogene (2000) 19, 1002-1009. Keywords: Wnt-1; breast cancer; mammary; transgenic; MMTV cancer models; Introduction Infection of most strains of mice, such as C3H, with mouse mammary tumor virus (MMTV) leads to a high incidence of mammary tumors (reviewed by Nusse, 1991). The sites of insertions by MMTV proviruses have been extensively mined in order to identify genes that are deregulated to cause tumorigenesis. Wnt-] was the first protooncogene to be cloned following activation by viral insertion in mouse mammary tumors (Nusse and Varmus, 1982). (Its initial name, int-1, was subsequently changed to Wnt-1 because of its homology to the Drosophila Wingless (Wg) gene (Nusse et al., 1991)). Insertional activation of Wnt-] occurs in approximately 70% of C3H mice that are chronically infected with MMTV (Nusse and Varmus, 1982). Other candidate protooncogenes that are some- times activated by MMTV proviral insertions include two additional members of the Wnt family, Wnt-3 (Roelink et al., 1990) and Wnt-10b (Lee et al., 1995); three members of the fibroblast growth factor family, Fegf-3/int-2 (Dickson et al., 1984), Fef-4/hst (Peters et al., 1989), and Fef-8/AIGF (MacArthur et al, 1995); Notch-4/int-3 (Lee et al., 1995); and int-6 (Asano et al., 1997), encoding a subunit of the translation initiation factor eIF3. Some of these genes, such as Fgf-3 and Wnt-10b, have been validated as oncogenes by *Correspondence: Y Li transgenic expression (Kwan et al., 1992; Lane and Leder, 1997; Muller et al., 1990). The Wnt-1 gene encodes a member of a large family of secreted proteins that are cysteine-rich, glycosylated, and poorly soluble (reviewed by Nusse and Varmus, 1992). Presently, at least 18 distinct Wnt family members have been identified in mammals. Having a propensity to associate with the extracellular matrix, Wnts act on both Wnt-producing and adjacent cells through cell surface receptors to control cell fate and patterning (reviewed by Nusse and Varmus, 1992). In mice, Wnt-I is expressed exclusively in the developing central nervous system (CNS) and aduit testes (Jakobovits et al., 1986; Shackleford and Varmus, 1987; Wilkinson et al., 1987), and it is required for CNS patterning and development of the midbrain and cerebellum (McMahon and Bradley, 1990; Thomas and Capecchi, 1990). Its Drosophila ortholog, Wg, controls segment polarity and many other developmental processes (Wodarz and Nusse, 1998). A brief overview of the Wnt signaling pathway One of the major intracellular responses to Wnt-1 signaling is to stabilize and increase the level of cytosolic f-catenin (Figure 1), a multi-functional protein that associates with membrane-bound E- cadherin, as well as several DNA binding proteins, such as members of the TCF/LEF family (reviewed by Kinzler and Vogelstein, 1996). Heterodimers of - catenin and transcriptions factors translocate to the nucleus and transactivate a number of genes, including c-myc (He et al., 1998), cyclin D1 (Shtutman ef ai., 1999; Tetsu and McCormick, 1999), WISPs (Pennica et al., 1998), and possibly cyclooxygenase-2 (Howe et al., 1999). Depending upon the cell type, Wnt signaling activates different genes, affecting various stages of development and several types of cancer. The receptors for Wnts have been identified as a class of seven transmembrane proteins known as Frizzled (Fz) (Bhanot et al, 1996). The ligand- receptor interaction is facilitated by extracellular proteoglycans and, inhibited by Fz-related proteins, dickkopf, and cerberus. After binding a Wnt ligand, Fz transmits a signal to cytoplasmic phosphoproteins in the disheveled (Dvl) family via unknown mechan- isms. Dvl1 inhibits the constitutively active kinase activity of glycogen synthase kinase type 3 (GSK3), which normally phosphorylates f-catenin and targets it for degradation. Cytosolic levels of B-catenin are additionally regulated by adenomatous polyosis coli (APC), which targets B-catenin for proteosome- mediated degradation, and by another large protein, Axin, also an inhibitor of Wnt signaling. A complex containing APC, Axin, B-catenin, GSK3, and GSK- ‘binding protein/Frat-1 has been observed in lysates prepared from certain cell types (reviewed by Barish and Williams, 1999). / Several members of the Wnt family transform cultured cells, when overexpressed. For example, overexpression of Wnt-1, Wnt-2, Wnt-3, and Wnht-3a (but not Wnt-4, Wnt-5a, Wnt-5b, and Wnt-7b) leads to morphological transformation of mammary epithelial cells such as CS7MG (Brown et al., 1986; Shimizu et al., 1997). Continued overexpression is required for the transformation phenotype induced by Wat-/ (Li et al., 1999; Mason et al., 1992). Wnt-I is not normally expressed in the mammary gland, nor has it been directly implicated in human breast cancer. However, several other Wnt family members are expressed in breast tissue, and some are overexpressed in breast tumors (reviewed by Bergstein and Brown, 1999). In addition, genes encoding several components and targets of the Wnt signaling pathway, including f-catenin, APC, E-cadherin, cyclin D1, c- myc, and WISPs, have been found to be mutated or deregulated in several types of human tumors, such as breast cancer (Bieche et a/., 1999), colon cancer (He et al., 1998), melanoma (Rimm et al., 1999; Rubinfeld et al., 1997), hepatocellular carcinoma (de La Coste et al., 1998), and pilomatricoma (Chan et al., 1999). The Wnt-1 transgenic (TG) mouse model Wnt-1 TG mice were initially made to test the oncogenicity of Wnt-1 (Tsukamoto et al., 1988). The transgene (Figure 2), is controlled by the Wht-1 promoter and an MMTV LTR inserted upstream of the gene in the opposite transcriptional orientation, in a fashion reminiscent of a typical viral insertion into the Wnt-1 locus in MMTV-induced tumors. Ectopic Wnt-I expression exerts a potent mitogenic effect on mammary epithelium; ductal hyperplasia is noticeable in the mammary end-buds by 18 days of gestation (Cunha and Hom, 1996) and very apparent 2 weeks after birth in the TG females (Lin et al., 1992). Because of the extensive ductal hyperplasia, female TG mice can not deliver milk to their young. About 50% of virgin female Wnt-1 TG mice in the SJL strain develop adenocarcinomas by 6 months of Nuclear Membrane v Target Genes Figure 1 Illustration of the Wnt signal transduction pathway (kindly provided by J-M Li). See text for explanation MMTV-Wnt-1 transgenic model Y Li et al age; the rest succumb to tumors by 1 year. Breeding females develop tumors slightly earlier than virgin mice (Shackleford e¢ al., 1993; Tsukamoto et al., 1988). This acceleration may be caused by either hormonal influence on cell growth or the. increased mass of the mammary epithelium attributed to pregnancies and lactation. Hyperplasia is also extensive in the primary mammary glands of adult male TG mice; about 15% of them develop palpable mammary tumors by | year of age (Kwan et al., 1992; Tsukamoto et al., 1988). Although metastasis does not seem to occur frequently at the time mammary tumors are detected (Tsukamoto et al., 1988), the majority of female Wnt-1 TG mice develop lymph node and/or jung metastasis after removal of the primary tumor (L Godley and WP Hively, unpublished observation). Tumors found in Wnt-l TG mice are usually moderately differentiated and comprised of ducts with multiple layers of epithelial cells, that show significantly higher than normal nucleus-to-cytoplasm ratio and occasionally pleomorphic nuclei and mitotic figures. The lumens usually contain pyknotic cells suggestive of apoptosis. Widespread necrosis and hemorrhage are sometimes noticeable in these tumors. In addition, extensive fibrosis is present in neoplasms induced by the Wnt-1 transgene. Hyperplastic glands of Wnt-1 TG mice also display a prominent fibrotic response, which may start as early as 7 days postnatally in TG females (G Cunha, personal communication). Variations in genetic backgrounds usually do not influence the time course of tumor development mediated by the Wnt-/] transgene (Bocchinfuso et al., 1999; Donehower et al., 1995; Shackleford et al., 1993; Tsukamoto et al., 1988). The original TG line was made in C57BL/6 X SJL F1 mice. Subsequently, interbreedings with other strains (FVB/N, BALB/c, 129/J, CS58BL/6) have been found to be similar to SLJ in tumor latency (Tabie 1). But a much longer latency has been observed in some mixed backgrounds (C Alexander, personal communication). CEE] LR es wiv LTR Wnt-1 > Wnet M SV40 poly A+ promoter signal Figure 2. Wnt-/ transgene construct. The 7 kb transgene contains the MMTV-LTR approximately 1 kb upstream of the mouse Wnt-] gene. The MMTV-LTR was placed in the opposite transcriptional orientation and is used as an enhancer. The Wnt-1 coding sequences are shown as filled boxes. A fragment containing the SV-40 splice and polyadenylation sites (850 bp) was placed downstream of the last exon of Wnt-1 Table 1 Genetic lesions crossed to the MMTV-Wat-/ transgene Genetic Genotype background MMTV-Fef-3 TG FVB/N References Muller e¢ al., 1990; Kwan et al., 1992 Sky —/- 129/Sv x CS7TBL/6 Lu et al., 1999; WP Hively (unpublished) ps3 —/- 129/Sv Donehower e7 al., 1992, 1995 ERa—/{- C57BL/6 Lubahn et al., 1993; Bocchinfuso et al., 1999 MMTV-TGF CS57BL/6 A Chytil, Y-L Chen and HL Moses (unpublished) 1003 Oncogene MMTV-Wnt-1 transgenic model Y Li et af 1004 Figure 3 Histology of mammary glands from an 18-week-old MMTV-Wnt-1 TG virgin female. (a) Hyperplastic mammary gland (25 x). (b) Mammary adenocarcinoma (25 x ) Many genetic lesions and epigenetic changes, such as levels of mammogenic hormones, have been implicated in breast carcinogenesis. However, the complex molecular interplay leading to breast cancer is very poorly understood. All mammary epithelial ceils expressing the transgene in the Wnt-1 TG line are at risk for tumor development. Indeed, ductal hyperplasia occurs throughout the mammary tissue early in development, yet tumors appear stochastically after several months. Therefore, other cooperative events must have accompanied expression of the Wht-1 transgene in the few cells that expanded into tumors. A number of methods have been applied to uncover these synergistic events. Among them are hormonal manipulations, insertional activation of protoonco- genes using retroviral infection, and breeding with TG and knockout mice carrying other genetic lesions implicated in breast cancer. Hormonal and stromal influences on Wnt-1-induced hyperplasia and tumors Estrogen is essential in mammary development and plays a very important role in carcinogenesis of the breast (reviewed by Pike ef al., 1993). It stimulates ductal morphogenesis and branching through nuclear receptors—ERa and possibly the recently identified ER (Kuiper et ai., 1996; Mosselman et al., 1996). ERa is expressed in both mammary epithelium and the stroma (Daniel ef al., 1987). ERB is detectable in mammary tissues at low levels, but its role in mammary proliferation remains elusive (Krege et al., 1998). Oncogene Hyperplastic ductal growth in the Wnt-1 TG animals persists despite estradiol deprivation due to ovariect- omy (Bocchinfuso et al., 1999; Lin et al., 1992). Albeit delayed, tumors continue to form in ovariectomized mice (Bocchinfuso et al., 1999), suggesting that Wnt-1 does not require estrogen signaling for stimulating proliferation and inducing tumors. These results were confirmed and strengthened by experiments in which the MMTV-Wnzt-/ transgene was crossed into the ERa knockout (ERKO) mice. In homozygous ERKO mice, mammary glands are underdeveloped, with rudimen- tary ducts confined to the nipple area (Lubahn et ai., 1993). The presence of the Wnt-J transgene stimulated hyperplastic ductal growth in ERKO mice, and the females developed mammary tumors at twice the age of Wnt-1 TG females with one or two intact ERa genes (Bocchinfuso et al., 1999). It remains to be determined if the increased latency to tumor development observed in ovariectomized or ERKO mice is the result of reduced mass of mammary epithelium, the loss of cooperative functions of ER signaling in Wht-I- induced oncogenesis, or both. The majority of human breast tumors are ERa- positive and respond to anti-hormone therapy; however, most malignant tumors are ERa-negative (McGuire and Clark, 1985). Together with the fact that only a small percentage of mammary epithelial cells express ERa (Petersen et al., 1987; Ricketts et al, 1991), it has been suggested that breast cancer may initiate from ER-positive cells but become ER-negative and estrogen-independent in its growth at later stages (Moolgavkar et al., 1980). The observation that mammary tumors arise in both ERKO and ovariecto- mized mice supports an alternative model that a fraction of breast cancers may directly evolve from ERa-negative cells (Nandi et al., 1995), an idea that needs to be tested with other oncogenic transgenes. The potent mitogenic effect of Wnt-1 on mammary epithelial cells may not depend upon other mammo- genic hormones either. For example, a similar degree of abnormal side branching was observed in mammary epithelial transplants derived from Wat-1 TG mice that were either wild-type or nullizygous for progesterone receptor « (PRa, C Brisken and R Weinberg, personal communication). Likewise, in ovariectomized and/or adrenalectomized mice, Wnt-1 continued to stimulate hyperplastic growth in transplanted and reconstituted glands (Edwards et al., 1992; Lin et al., 1992). The reciprocal interactions between parenchyma and stroma are important in mammary development, remodeling, and carcinogenesis (reviewed by Cunha and Hom, 1996). For example, signaling through the epidermal growth factor receptor (EGFR) in the mesenchyme is required for ductal growth and branching morphogenesis, since epithelium trans- planted from wild-type mice fails to proliferate in the fat-pad from EGFR-null mice (Wiesen et al., 1999). Interestingly, this requirement also seems to be diminished in Wnt-1 TG mice. Transplantation of epithelium from Wnt-] TG animals into the fat-pad of EGFR nullizygous mice only modestly impaired hyperplastic growth (G Cunha, personal communica- tion). Epithelial-stromal interactions in tumor forma- tion have also been studied by experiments in which mammary epithelial cells from Wnt-1 TG animals were transplanted into rat mammary fat-pad. Transplanta- tion led to fibrotic proliferation in rat mesenchyme (G Cunha, personal communication), suggesting that the alteration of stromal differentiation is mediated by the Wnt-1-expressing epithelial cells. Wnt-mediated epithe- lial-mesenchymal interactions have also been reported . in other tissues. For example, Wnt-induced mesench- ymal reactions may regulate axonal growth and guidance in developing limbs. Several members of the Wnt family expressed in limb ectoderm induce production of neurotrophin-3 in the underlying mesenchyme (Patapoutian et al., 1999). Collaboration between Wnt-1 and other genes in oncogenesis ‘Mammary tumors induced by MMTV occasionally show transcriptional activation of both Wat-1 and fibroblast growth factor3 (Fgf3, Peters et al., 1986), suggesting that these two genes collaborate in oncogenesis. Fgf3 belongs to a family of heparin- binding proteins that are both mitogenic and angio- genic. Signaling by FGFs is mediated by transmem- brane receptors (FGFRs) that phosphorylate and activate several substrates, leading to the activation of mitogen-activated protein kinases (reviewed by Faham et al., 1998). Although Wnts and FGFs act through very different pathways, they are both required for development of primary body axis, neural axis, limbs, and other structures, suggesting that these two families of growth factors may collaborate in develop- ment in ways that resemble synergistic roles in tumor formation. Transgenic female mice expressing MMTV-Fe/f3 show extensive mammary hyperplasia but rarely develop tumors (Muller et al., 1990). When this transgene was bred into Wni-1 TG mice, tumors developed faster in bi-transgenic females than in females bearing either transgene alone, providing direct evidence of cooperation between these two growth factors (Kwan et al., 1992). The acceleration is even more dramatic in the bi-transgenic males. Additional evidence of synergistic interactions between Wnt-/ and members of the Fgf family comes from infection of Wnt- 1 TG animals with MMTV (Shackleford ef al., 1993). Infection accelerates tumor formation, and up to ten tumors per mouse were observed in infected animals. Approximately 40% of the mammary tumors showed insertional activation of Fgf3, a small percentage of them had insertional activation of both Fgf3 and Fgf-4 or Fgf4 alone. Another member of the Fgf family, Fgf-8, was also found to be insertionally-activated and/or overexpressed in some of these tumors (Kapoun and Shackleford, 1997; MacArthur et al., 1995). Collabora- tion between members of the Wnt and FGF families has also been observed in experiments in which infection of MMTV-Fef3 TG mice with MMTV led to frequent viral insertions in Wnt-1 or Wnt-10b loci (Lee et al., 1995). Tumor growth factor £ (TGF) stimulates cell growth under some conditions, but, more commonly, inhibits celi proliferation, especially in the mammary gland (reviewed by Massague, 1998). For example, transgenic expression of TGF inhibits tumor forma- tion in mice expressing an MMTV-7GF«e transgene (Pierce et al., 1995). But in a recent cross between our MMTV-Wni-I TG mice and MMTV-TGFB TG animals, no éffects were observed on the rate of MMTV-Wnt-1 transgenic model Y Li et al tumor appearance, histology, or the size of the tumor induced by the Wnt-] transgene (A Chytil, Y-L Chen and HL Moses, personal communication). Assuming adequate levels of expression, it appears TGF cannot inhibit proliferation of mammary epithelia stimulated by the Wnt-/ transgene. Collaboration between Wnt-1 and loss of a tumor suppressor gene Several tumor suppressor genes are mutated or downregulated in human breast cancer. Inherited mutations of some of them predispose to breast neoplasm. For example, mutations of BRCA-1 and BRCA-2 are found in approximately 50% and 30%, respectively, of families predisposed to breast cancer ‘(Ford et al., 1998). Somatic p53 mutations are found in about 35% of sporadic and 85% of familial breast cancers (Crook et al., 1998), and germline alterations of p53 are associated with a predisposition to several cancers, including breast cancer (the Li-Fraumeni syndrome). RB, a cell cycle regulator, is also mutated in a small percentage of sporadic human breast cancers (Berns et al., 1995). In addition, p21/WAFI/ CIPI, a_ cyclin-dependent kinase inhibitor that regulates Gi-S cell cycle progression, is downregu- lated in some breast tumors, especially those with poor prognosis (Jiang et al., 1997; Wakasugi et al., 1997). The impact of the loss of a tumor suppressor gene on tumorigenesis has been documented in animal modeis using targeted gene disruption, loss of heterozygosity (LOH) assays, and transgenic overexpression of a dominant-negative version of a tumor suppressor gene. Mice deficient for the p53 tumor suppressor gene (p53+/— and p53—/—) develop tumors of non- epithelial origin (Donehower et al., 1992). To analyse the effect of p53 inactivation on mammary oncogenesis, p53 knockout mice were bred with Wnt-1 TG mice (Donehower et al., 1995). p53 nullizygotes (both females and males) expressing the Wnt-] transgene develop mammary tumors much earlier than mice containing at least one wild-type allele, suggesting that inactivation of p53 plays an important role and collaborates with Wnt- J in mammary oncogenesis. In addition, p53-null tumors are more anaplastic and less fibrotic than tumors that carry at least one copy of the p53 gene (Donehower et al., 1995). Although the absence of one copy of p53 did not significantly alter the time at which MMTV-Wnt-/ transgene induced tumors appeared, approximately 50% of the tumors in p53-heterozygous, Wnt-1 TG mice displayed loss of the wild-type locus. This frequent occurrence of LOH contrasts with the very rare loss of the wild-type p53 allele in mammary tumors from p53 heterozygotes carrying an MMTV-c- myc transgene (Elson et al., 1995; McCormack et al., 1998). It is notable that inactivation of p53 collaborates with MMTV-c-myc, MMTV-H-ras, and MMTV-neu transgenes to produce lymphomas and salivary tumors, but rarely mammary tumors (C-X Deng, personal communication, Elson et al., 1995; Hundley ef al. 1997). Mutations in. BRCA-I and BRCA-2 are often associated with loss of p53 in breast carcinogenesis in humans (Crook et al., 1998). Induction of mammary 1005 Oncogene MMTV-Wnt-1 transgenic model Y Li et al 1006 tumors in the mouse by mammary-specific Brcal inactivation is dramatically accelerated by inactivation of p53 (Xu et al., 1999). However, loss of one allele of Brca-1 (T Wynshaw-Boris, personal communication) or Brca-2. (XS Cui and LA _ Donehower, personal communication) does not seem to influence the kinetics of tumor formation induced by the Wnt-1 transgene. It remains to be determined whether tumors that are heterozygous for Brcal or Brca2 show LOH or alteration in karyotype. The availability of mice that carry loxP-flanked (floxed) alleles of Brcal and Brca2 will permit better tests for synergy between the loss of Brcal or Brca2 and inheritance of a Wnt-] transgene in tumor formation. Germline mutations in one copy of the Rb gene predispose humans to retinoblastomas and osteosarco- mas. Mice nullizygous for Rb die during embryogen- esis; heterozygotes develop tumors primarily in the pituitary and thyroid glands but rarely in mammary glands (Jacks et al., 1992). To determine if the loss of Rb affects the development of tumors in Wnat-1 TG animals, we have crossed Wnt-/ TG animals with mice heterozygous for Rb. Absence of one allele of Rb did not affect the age at which the tumor was detected, and none of 25 tumors examined by restriction mapping showed loss of the wild-type locus (WP Hively, unpublished). The lack of acceleration may be due to the complementary expression of one or both of the other two members of the Rb gene family (p/07 and p130) in the mouse mammary gland. In fact, the presence of normal p/07 aileles has been shown to inhibit Rb deficiency-mediated tumor formation in the mouse retina (Robanus-Maandag ef al., 1998). Elim- ination of all three members of the Rb gene family in Wnt-I1 TG mice would further clarify the role of their inactivation in mammary oncogenesis. One way to eliminate their functions is to generate transgenic mice expressing the gene encoding the amino terminal domain (T,,;) of the Simian virus 40 T antigen, which inactivates all three members of the Rb family (Saenz Robles et al., 1994; Symonds ez al., 1994). Inactivation of one or both alleles of p2/ did not accelerate tumor formation in Wnt-1 TG mice (Jones et al., 1999). But, interestingly, tumors from p2/+/— mice grew significantly faster, with a higher mitotic index and increased cyclin D1-associated phosphoryla- tion of Rb, than those from either p2/+/+ or p21—/— mice (Jones et al., 1999). Additional tumor suppressor genes that collaborate with the Wnt-] transgene to induce tumor formation may be identified by scanning the whole genome for LOH. Application of this approach has led to the identification of two regions on mouse chromosomes 9 and 16 that are frequently deleted in insulinomas and carcinoid tumors in TG mice expressing the Simian virus 40 large T antigen (Dietrich et al., 1994). Furthermore, using this technology in Fl hybrid mice between FVB/N and Mus musculus castaneus, Radany and colleagues (1997) have found that a marker on chromosome 4 from Mus musculus castaneus was frequently lost in MMTV-H-Ras transgene-induced mammary tumors. In contrast, no single chromosome was preferentially lost in tumors occurring in Fl progeny of a similar cross between Wnt-1 TG SJL mice and Mus musculus castaneus (unpublished data of K Hong et al., cited in Radany et al., 1997). Oncogene Molecular characterization of tumors from Wnt-1 TG animals Chromosomal rearrangements including aneuploidy, chromosomal translocations and duplications, and amplification of selected genes are common in tumor cells (reviewed by Wright, 1999). Loss of p53 function frequently leads to deregulated ceil cycle control and chromosomal instability, which favors tumor growth (reviewed by Prives and Hall, 1999). Mammary tumors from Wnt-1 TG mice with one or two functional copies of p53 display occasional chromosomal abnormalities as shown by comparative genome hybridization (CGH, Kallioniemi et al., 1992), which detects regions of expansion and deletion in all chromosomes. As expected, Wnt-1 induced tumors without any p53 function usually have more than one chromosomal abnormality. Tumors that arose in p53 heterozygotes and experienced LOH at the p53 locus displayed even more extensive alterations (at least three regions of DNA gain or loss) (Donehower et al., 1995). In general, it is difficult to anticipate what specific genes in an amplified chromosomal region may have synergized with an oncogenic transgene to induce neoplasm. But the distal region of chromosome 7, which was amplified in a Whnt-I-induced p53—/— tumor, is the site of Fgf3 (Donechower et al., 1995). Molecular hybridization using an Fgf3-specific probe confirmed that Ffg3 was amplified and abundantly expressed in this tumor (Donehower et al., 1995). This is different from human breast cancer, in which the syntenic region of chromosome 8q is frequently amplified (Brison, 1993; Lammie et al., 1991; Theiliet et al., 1989), but Fgf3 mRNA is not detected (Penault- Llorca et al., 1995); however, a linked gene, PRAD-1/ CycinD1, is usually overexpressed in such tumors (Motokura et al., 1991). Spectral karyotyping (SKY) labels each chromosome with a different color, allowing detection of chromo- somal translocations and duplications (Liyanage et al., 1996). We have analysed some tumors from Wnt-1 TG mice that were p53+/— or p53—/—. Translocations, trisomy, and aneuploidy have been detected in cells cultured from some of these tumors (Z Weaver and WP Hively, unpublished). Karyotype instability in mammary tumors has been reported in mammary- specific Brcal knockout mice (Xu et al., 1999) and other transgenic models including MMTV-c-myc (McCormack et al., 1998; Weaver et al., 1999). As a physiologic response to genotoxins, p53 is rapidly induced to cause cell cycle arrest and/or apoptosis. Inactivation of p53 is often accompanied by accelerated cell growth and attenuated apoptosis (reviewed by Ko and Prives, 1996). p53 deficiency (p53+/—, p53—/—) enhances cell proliferation in the Wnt-1 transgene-derived tumors, but the modestly ongoing apoptosis that accompanies Wnt-1 overexpres- sion does not seem to be attenuated (Jones e¢ al., 1997). Similarly, absence of one allele of p53 does not affect the apoptotic index in mammary tumors induced by an MMTV-c-myc transgene (McCormack et al., 1998). Normal telomeres are essential to cell survival. Telomerase is usually activated in human cancer cells, presumably to overcome shortened telomeres due to excessive cell replication (reviewed by de Lange and DePinho, 1999). Normal telomeres (20-50 kb) are present in both hyperplastic glands and carcinomas from Wnt-1 TG mice, regardless of the p53 status (Broccoli et al., 1996). Interestingly, despite the presence of long telomeres, telomerase activity and the RNA component of the enzyme were consistently upregulated in these tumors compared with normal and hyperplastic glands (Broccoli et al., 1996), suggesting that activation of the telomerase machinery in at least some mammary tumors does not depend upon telomeric shortenings. Breeding Wnt-1 TG animals with mice that carry a mutated gene for telomerase or a component of the telomeric complex (Blasco et al., 1997; Rudolph et al., 1999) will help address whether the telomere activation is required for mammary oncogenesis induced by the Wnt-/ transgene. Another approach to uncovering the molecular basis of tumorigenesis is to identify differentially- expressed genes during various stages of tumor formation. Several methods including subtractive hybridization, differential display, serial analysis of gene expression (SAGE), and cDNA expression array technology have been used. A number of genes have been found to be deregulated in Whnt-1-induced tumors and Wnt-I-transformed cells by these and other methods. For example, using PCR to screen for differentially expressed tyrosine kinases, we found that Sky, which encodes a member of the Axl/Ufo family of receptor tyrosine kinases, is barely detectable in the mammary glands from virgin animals and in preneoplastic mammary glands, but is abundantly expressed in the mammary tumors of Wnat-1 TG mice (Taylor et al., 1995). Recently, using a modified subtractive hybridization approach, Pennica et al. (1998) have found that two novel genes, WISP] and 2, are overexpressed in Wnt-J-transformed mammary cells and that they are transcriptionally regulated by Wnt-I expression and aberrantly expressed in colon cancer. Screening for upregulated genes in tumors might also help identify collaborating factors in tumor formation. But alteration of the transcriptional apparatus during neoplastic conversion may dereg- ulate many non-collaborating genes. An example of such a non-synergistic element is Sky. Tumor development in Whot-] transgenic mice was not affected by breeding Sky knockout mice (Lu et al., 1999) with Wnt-1 TG animals (WP Hively, unpub- lished), suggesting that overexpression of Sky is not necessary for Wnt-1 mediated oncogenesis. Involvement of other components of the Wnt signaling pathway in mammary carcinogenesis Many other components of the Wnt signaling path- way have been implicated in mammary tumorigenesis. Mutations in APC, a negative regulator of the Wnt signaling pathway, have been reported to confer an increased risk for development of breast cancer in Ashkenzi Jews (Redston et al., 1998; Woodage et al., 1998). Min mice, which carry a nonsense mutation at one APC locus, also have increased risk for mammary carcinomas after carcinogen treatment (Moser et al., 1993, 1995). With the use of additional TG mice, it will be interesting to determine if deregulated expression of other components of the Wont-1 signaling pathway, such as over-expression of f- MMTV-Wnit-1 transgenic model Y Li et af catenin and inactivation of &-cadherin, also induce mammary tumors. Ali three members of the Dv/ family (Dv/1, Dvi2 and Dvi3) are expressed in mammary glands, with Dvll being most abundant (Tsang et al., 1996). Dvi proteins transmit signals from the Fz receptor for Wnt-1 to B- catenin via unknown mechanisms (see Introduction). Mice nullizygous for Dvil are normal except for abnormalities in social behavior and sensorimotor gating (Lijam et ai., 1997). The mammary epithelia lacking the dominant member of this family might be expected to respond poorly to Wnt-1 induced cell proliferation and tumor formation. But, Dv// nullizyg- osity did not affect the rate of tumor formation in Wnt- I TG mice (N Lijam, WP Hively, HE Varmus and T Wynshaw-Boris, unpublished). Since Dv/2 and Dvi/3 are also expressed in the mammary gland, they might have substituted for Dv// in mediating the Wnt-1 signal. Syndecan-1 is a member of the transmembrane proteoglycan family that regulates ceil morphology and growth (Leppa et al., 1992). Proteoglycans facilitate the binding of Wnt ligands to Fz receptors (Lin and Perrimon, 1999; Wodarz and Nusse, 1998). Consistent with this finding, Wnt-1 TG mice that carry two null alleles of syndecan-] very rarely develop tumors (C Alexander, personal communication), suggesting that syndecan-1 may be an important factor in mediating Wnt-1 signaling in the mammary gland. Prospects Although initially developed to document the onco- genic potential of Wnt-1, our line of MMTV-Wnt-1 TG mice has been useful in studying many aspects of mammary tumorigenesis: the cooperation between cancer genes, the influence of estrogen receptors and growth hormones, and the concomitant changes in genomic instability and gene expression. Different stages of tumor progression can be discerned in Wnt-1 TG mice, and some of the collaborative lesions accompanying Wnt-1 overexpres- sion in tumor formation have been defined. There- fore, this line may be a convenient source of hyperplastic glands and invasive and metastatic tumors for various approaches designed to identify molecular signatures of tumor progression. Compar- ing the expression profile of the Wnt-J-derived tumors with those of tumors derived from Wat-1 TG mice crossed with other genetically modified lines may offer additional insights into the complex nature of mammary oncogenesis. Additional benefits in the characterization of these tumors include identification of transcriptional targets of the Wnt-1 signaling pathway. Recently, a novel method has been used to transduce oncogenes into somatic cells of a specific tissue (reviewed by Fisher et al., 1999) using sub-group A avian leukosis virus (ALV-A) as a vector. Transgenic expression of tv-a, encoding the receptor for ALV-A, from a cell type-specific promoter, permits tissue- specific infection with ALV-A, which does not produce infectious virus in mammalian hosts. Conse- quently, combinatorial effects of genetic lesions can be examined in a single TG line by infecting with mixtures of ALV-A viruses expressing different oncogenes. In addition, ALV-A expressing the gene encoding Cre 1007 Oncogene MMTV-Wnt-1 transgenic model Y Li et al 1008 recombinase can be used to inactivate tumor suppres- sor genes flanked with loxP recombination sites. Since ALV infection requires mitotic cells which are widely available only during late pregnancy, breeding the Wnt-1 transgene into mice expressing tv-a from a mammary-specific promoter may provide both replicat- ing epithelial cells (eliminating the requirement for pregnancies) and a cancer predisposing factor allowing more rapid formation of tumors. The TVA technology References Asano K, Merrick WC and Hershey JW. (1997). J. Biol. Chem., 272, 23477 —23480. Barish GD and Williams BO. (1999). Signal Networks and Cell Cycle Control. Gutkind JS (ed.). Human Press. Bergstein I and Brown AMC. (1999). Breast Cancer: Molecular Genetics, Pathogenesis and Therapeutics. Bow- cock AM (ed.). Human Press: Totowa, New Jersey, pp. 181-198. Berns EM, de Klein A, van Putten WL, van Staveren IL, Bootsma A, Klijn JG and Foekens JA. (1995). Int. J. Cancer, 64, 140-145. Bhanot P, Brink M, Samos CH, Hsieh JC, Wang Y, Macke JP, Andrew D, Nathans J and Nusse R. (1996). Nature, 382, 225-230. Bieche I, Laurendeau I, Tozlu 8, Olivi M, Vidaud D, Lidereau R and Vidaud M. (1999). Cancer Res., 59, 2759-2765. Blasco MA, Lee HW, Hande MP, Samper E, Lansdorp PM, DePinho RA and Greider CW. (1997). Cell, 91, 25-34. Bocchinfuso WP, Hively WP, Couse JF, Varmus HE and Korach KS. (1999). Cancer Res., 59, 1869 — 1876. Brison O. (1993). Biochim Biophys Acta, 1155, 25-41. Broccoli D, Godley LA, Donehower LA, Varmus HE and de Lange T. (1996). Mol. Cell Biol., 16, 3765-3772. Brown AM, Wildin RS, Prendergast TJ and Varmus HE. (1986). Cell, 46, 1001-1009. Chan EF, Gat U, McNiff JM and Fuchs E. (1999). Nat. Genet., 21, 410-413. Crook T, Brooks LA, Crossland S, Osin P, Barker KT, Waller J, Philp E, Smith PD, Yulug I, Peto J, Parker G, Allday MJ, Crompton MR and Gusterson BA. (1998). Oncogene, 17, 1681 — 1689. Cunha GR and Hom YK. (1996). J. Mammary Gland Biol. Neoplasia, 1, 21—37. Daniel CW, Silberstein GB and Strickland P. (1987). Cancer Res., 47, 6052-6057. de La Coste A, Romagnolo B, Billuart P, Renard CA, Buendia MA, Soubrane O, Fabre M, Chelly J, Beldjord C, Kahn A and Perret C. (1998). Proc. Natl. Acad. Sci. USA, 95, 8847-8851. de Lange T and DePinho RA. (1999). Science, 283, 947-949. Dickson C, Smith R, Brookes S.and Peters G. (1984). Cell, 37, 529-536. Dietrich WF, Radany EH, Smith JS, Bishop JM, Hanahan D and Lander ES. (1994). Proc. Natl. Acad. Sci. USA, 91, 9451-9455. Donehower LA, Godley LA, Aldaz CM, Pyle R, Shi YP, Pinkel D, Gray J, Bradley A, Medina D and Varmus HE. (1995). Genes Dev., 9, 882-895. Donehower LA, Harvey M, Slagle BL, McArthur MJ, Montgomery CA, Butel JS and Bradley A. (1992). Nature (London), 356, 215-221. Edwards PA, Hiby SE, Papkoff J and Bradbury JM. (1992). Oncogene, 7, 2041-2051. Elson A, Deng C, Campos-Torres J, Donehower LA and Leder P. (1995). Oncogene, 11, 181-190. Faham S, Linhardt RJ and Rees DC. (1998). Curr. Opin. Struct. Biol., 8, 578 —586. Oncogene may help test candidate collaborative events in context of the Wnt-1 transgene. Acknowledgments We thank S Orsulic, S Ortiz, G Fisher and M Chamorro for critical review of the manuscript. Y Li is supported by the Department of Defense Breast Cancer Research Program. Fisher GH, Orsulic S, Holland EC, Hively WP, Li Y, Lewis BC, Williams BO and Varmus HE. (1999). Oncogene, 18, 5253-5260. Ford D, Easton DF, Stratton M, Narod S, Goldgar D, Devilee P, Bishop DT, Weber B, Lenoir G, Chang-Claude J, Sobol H, Teare MD, Struewing J, Arason A, Scherneck S, Peto J, Rebbeck TR, Tonin P, Neuhausen S, Barkardottir R, Eyfjord J, Lynch H, Ponder BA, Gayther SA, Zelada-Hedman M et al. (1998). Am. J. Hum. Genet., 62, 676 —689. He TC, Sparks AB, Rago C, Hermeking H, Zawel L, da Costa LT, Morin PJ, Vogelstein B and Kinzler KW. (1998). Science, 281, 1509-1512. Howe LR, Subbaramaiah K, Chung WJ, Dannenberg AJ and Brown AM. (1999). Cancer Res., 59, 1572 —1577. Hundley JE, Koester SK, Troyer DA, Hilsenbeck SG, Subler MA and Windle JJ. (1997). Mol. Cell Biol., 17, 723-731. Jacks T, Fazeli A, Schmitt EM, Bronson RT, Goodell MA and Weinberg RA. (1992). Nature, 359, 295-300. . Jakobovits A, Shackleford GM, Varmus HE and Martin GR. (1986). Proc. Natl. Acad. Sci. USA, 83, 7806-7810. Jiang M, Shao ZM, Wu J, Lu JS, Yu LM, Yuan JD, Han QX, Shen ZZ and Fontana JA. (1997). Int. J. Cancer, 74, 529 — $34. Jones JM, Attardi L, Godley LA, Laucirica R, Medina D, Jacks T, Varmus HE and Donehower LA. (1997). Cell Growth Differ., 8, 829-838. Jones JM, Cui XS, Medina D and Donehower LA. (1999). Cell Growth Differ, 10, 213-222. Kallioniemi A, Kallioniemi OP, Sudar D, Rutovitz D, Gray JW, Waldman F and Pinkel D. (1992). Science, 258, 818 - 821. Kapoun AM and Shackleford GM. (1997). Oncogene, 14, 2985-2989. Kinzler KW and Vogelstein B. (1996). Cell, 87, 159-170. Ko LJ and Prives C. (1996). Genes Dev., 10, 1054-1072. Krege JH, Hodgin JB, Couse JF, Enmark E, Warner M, Mahler JF, Sar M, Korach KS, Gustafsson JA and Smithies O. (1998). Proc. Natl. Acad. Sci. USA, 95, 15677 — 15682. Kuiper GG, Enmark E, Pelto-Huikko M, Nilsson S and Gustafsson JA. (1996). Proc. Natl. Acad. Sci. USA, 93, 5925-5930. Kwan H, Pecenka V, Tsukamoto A, Parslow TG, Guzman R, Lin TP, Muller WJ, Lee FS, Leder P and Varmus HE. (1992). Mol. Cell Biol., 12, 147-154. Lammie GA, Fantl V, Smith R, Schuuring E, Brookes S, Michalides R, Dickson C, Arnold A and Peters G. (1991). Oncogene, 6, 439-444. Lane TF and Leder P. (1997). Oncogene, 15, 2133-2144. Lee FS, Lane TF, Kuo A, Shackleford GM and Leder P. (1995). Proc. Natl. Acad. Sci. USA, 92, 2268 —2272. Leppa S, Mali M, Miettinen HM and Jalkanen M. (1992). Proc. Natl. Acad. Sci. USA, 89, 932-936. Li YX, Papkoff J and Sarkar NH. (1999). Virology, 255, 138-149. Lijam N, Paylor R, McDonald, MP, Crawley JN, Deng CX, Herrup K, Stevens KE, Maccaferri G, McBain CJ, Sussman DJ and Wynshaw-Boris A. (1997). Cell, 90, 895-905. Lin TP, Guzman RC, Osborn RC, Thordarson G and Nandi S. (1992). Cancer Res., 52, 4413-4419. Lin X and Perrimon N. (1999). Nature, 400, 281-284. Liyanage M, Coleman A, du Manoir S, Veldman T, McCormack S, Dickson RB, Barlow C, Wynshaw-Boris A, Janz S, Wienberg J, Ferguson-Smith MA, Schrock E and Ried T. (1996). Nat. Genet., 14, 312-315. Lu Q, Gore M, Zhang Q, Camenisch T, Boast S, Casagranda F, Lai C, Skinner MK, Klein R, Matsushima GK, Earp HS, Goff SP and Lemke G. (1999). Nature, 398, 723-728. Lubahn DB, Moyer JS, Golding TS, Couse JF, Korach KS and Smithies O. (1993). Proc. Natl. Acad. Sci. USA, 90, 11162-11166. MacArthur CA, Shankar DB and Shackleford GM. (1995). J. Virol., 69, 2501 —2507. Mason JO, Kitajewski J and Varmus HE. (1992). Mol. Biol. Cell, 3, 521-533. Massague J. (1998). Annu. Rev. Biochem., 67, 753-791. McCormack SJ, Weaver Z, Deming S, Natarajan G, Torri J, Johnson MD, Liyanage M, Reid T and Dickson RB. (1998). Oncogene, 16, 2755 —~2766. McGuire WL and Clark GM. (1985). Semin. Oncol., 12, 12- 16. McMahon AP and Bradley A. (1990). Cell, 62, 1073-1085. Moolgavkar SH, Day NE and Stevens RG. (1980). J. Natl. Cancer Inst., 65, 559 —569. Moser AR, Luongo C, Gould KA, McNeley MK, Shoe- maker AR and Dove WF. (1995). Eur. J. Cancer, 31A, 1061-1064. Moser AR, Mattes EM, Dove WF, Lindstrom MJ, Haag JD and Gould MN. (1993). Proc. Natl. Acad. Sci. USA, 90, 8977-8981. Mosselman S, Polman J and Dijkema R. (1996). FEBS Lett., 392, 49-53. . Motokura T, Bloom T, Kim HG, Juppner H, Ruderman JV, Kronenberg HM and Arnold A. (1991). Nature, 350, 512-— 515. Muller WJ, Lee FS, Dickson C, Peters G, Pattengale P and Leder P. (1990). EMBO J., 9, 907-913. Nandi 8, Guzman RC and Yang J. (1995). Proc. Natl. Acad. Sci. USA, 92, 3650-3657. Nusse R. (1991). Curr. Top. Microbiol. Immunol., 171, 43 - 65. Nusse R, Brown A, Papkoff J, Scambler P, Shackleford G, McMahon A, Moon R and Varmus H. (1991). Cell, 64, 231. Nusse R and Varmus HE. (1982). Cell, 31, 99-109. Nusse R and Varmus HE. (1992). Cell, 69, 1073 — 1087. Patapoutian A, Backus C, Kispert A and Reichardt LF. (1999). Science, 283, 1180-1183. Penault-Llorca F, Bertucci F, Adelaide J, Parc P, Coulier F, Jacquemier J, Birnbaum D and deLapeyriere O. (1995). Int. J. Cancer, 61, 170-176. Pennica D, Swanson TA, Welsh JW, Roy MA, Lawrence DA, Lee J, Brush J, Taneyhill LA, Deuel B, Lew M, Watanabe C, Cohen RL, Melhem MF, Finley GG, Quirke P, Goddard AD, Hillan KJ, Gurney AL, Botstein D and Levine AJ. (1998). Proc. Natl. Acad. Sci. USA, 95, 14717— 14722. Peters G, Brookes S, Smith R, Placzek M and Dickson C. (1989). Proc. Natl. Acad. Sci. USA, 86, 5678 — 5682. Peters G, Lee AE and Dickson C. (1986). Nature, 320, 628 — 631. Petersen OW, Hoyer PE and van Deurs B. (1987). Cancer Res., 47, 5748-5751. Pierce Jr DF, Gorska AE, Chytil A, Meise KS, Page DL, Coffey Jr RJ and Moses HL. (1995). Proc. Natl. Acad. Sci. USA, 92, 4254-4258. MMTV-Wot-i transgenic model Y Li et af @ Pike MC, Spicer DV, Dahmoush L and Press MF. (1993). Epidemiol Rev., 15, 17-35. Prives C and Hall PA. (1999). J. Pathol., 187, 112-126. Radany EH, Hong K, Kesharvarzi S, Lander ES and Bishop JM. (1997). Proc. Natl. Acad. Sci. USA, 94, 8664-8669. Redston M, Nathanson KL, Yuan ZQ, Neuhausen SL, Satagopan J, Wong N, Yang D, Nafa D, Abrahamson J, Ozcelik H, Antin-Ozerkis D, Andrulis I, Daly M, Pinsky L, Schrag D, Gallinger S, Kaback M, King MC, Woodage T, Brody LC, Godwin A, Warner E, Weber B, Foulkes W and Offit K. (1998). Nat. Genet., 20, 13-14. Ricketts D, Turnbull L, Ryall G, Bakhshi R, Rawson NS, Gazet JC, Nolan C and Coombes RC. (1991). Cancer Res., §1, 1817-1822. Rimm DL, Caca K, Hu G, Harrison FB and Fearon ER. (1999). Am. J. Pathol., 154, 325-329. Robanus-Maandag E, Dekker M, van der Valk M, Carrozza ML, Jeanny JC, Dannenberg JH, Berns A and te Riele H. (1998). Genes Dev., 12, 1599-1609. Roelink H, Wagenaar E, Lopes da Silva S, and Nusse R. (1990). Proc. Natl. Acad. Sci. USA, 87, 4519-4523. Rubinfeld B, Robbins P, El-Gamil M, Albert I, Porfiri E and Polakis P. (1997). Science, 275, 1790-1792. Rudolph KL, Chang S, Lee HW, Blasco M, Gottlieb GJ, Greider C and DePinho, RA. (1999). Cell, 96, 701 —712. Saenz Robles MT, Symonds H, Chen J and Van Dyke T. (1994). Mol. Cell Biol., 14, 2686-2698. Shackleford GM, MacArthur CA, Kwan HC and Varmus HE. (1993). Proc. Natl. Acad. Sci. USA, 90, 740-744. Shackleford GM and Varmus HE. (1987). Cell, 50, 89-95. Shimizu H, Julius MA, Giarre M, Zheng Z, Brown AM and Kitajewski J. (1997). Cell Growth Differ, 8, 1349-1358. Shtutman M, Zhurinsky J, Simcha I, Albanese C, D’Amico M, Pestell R and Ben-Ze’ev A. (1999). Proc. Natl. Acad. Sci. USA, 96, 5522-5527. Symonds H, Krall L, Remington L, Saenz-Robles M, Lowe S, Jacks T and Van Dyke T. (1994). Cell, 78, 703-711. Taylor IC, Roy S, Yaswen P, Stampfer MR and Varmus HE. (1995). J. Biol. Chem., 270, 6872 — 6880. Tetsu O and McCormick F. (1999). Nature, 398, 422 —426. Theillet C, Le Roy X, De Lapeyriere O, Grosgeorges J, Adnane J, Raynaud SD, Simony-Lafontaine J, Goldfarb M, Escot C and Birnbaum D. (1989). Oncogene, 4, 915- 922. Thomas KR and Capecchi MR. (1990). Nature, 346, 847- 850. Tsang M, Lijam N, Yang Y, Beier DR, Wynshaw-Boris A and Sussman DJ. (1996). Dev. Dyn., 207, 253 —262. Tsukamoto AS, Grosschedi R, Guzman RC, Parslow T and Varmus HE. (1988). Cell, 55, 619-625. Wakasugi E, Kobayashi T, Tamaki Y, Ito Y, Miyashiro I, Komoike Y, Takeda T, Shin E, Takatsuka Y, Kikkawa N, Monden T and Monden M. (1997). Am. J. Clin. Pathol., 107, 684-691. Weaver ZA, McCormack SJ, Liyanage M, du Manoir S, Coleman A, Schrock E, Dickson RB and Ried T. (1999). Genes Chromos. Cancer, 25, 251-260. Wiesen JF, Young P, Werb Z and Cunha GR. (1999). Development, 126, 335-344. Wilkinson DG, Bailes JA and McMahon AP. (1987). Cell, 50, 79-88. Wodarz A and Nusse R. (1998). Ann. Rev. Cell Dey. Biol., 14, 59-88. Woodage T, King SM, Wacholder S, Hartge P, Struewing JP, McAdams M, Laken SJ, Tucker MA and Brody LC. (1998). Nat. Genet., 20, 62-65. Wright EG. (1999). J. Pathol., 187, 19-27. Xu X, Wagner KU, Larson D, Weaver Z, Li C, Ried T, Hennighausen L, Wynshaw-Boris A and Deng CX. (1999). Nat. Genet., 22, 37-43. 009 Oncogene