IIIIMWWWHIHIWIIHWIHHIWHIIHWIHWI This is to certify that the thesis entitled MASPIN, A TUMOR SUPPRESSOR GENE, IS EXPRESSED IN HUMAN BASAL BREAST EPITHELIAL CELLS BUT NOT IN BREAST EPITHELIAL STEM CELLS AND BREAST CARCINOMA CELLS presented by Maki Saitoh has been accepted towards fulfillment of the requirements for the MS. degree in Genetics Program (Ck-C nmj (4‘é , Major Professor’s Signature Ja :1 .Q 2 8 1 2 6 0 7L / Date MSU is an Affirmative Action/Equal Opportunity Institution LIBRARY Michigan State University PLACE IN RETURN BOX to remove this checkout from your record. To AVOID FINES return on or before date due. MAY BE RECALLED with earlier due date if requested. DATE DUE DATE DUE DATE DUE 6/01 cJCIRC/DatoDuopss-sz MASPIN, A TUMOR SUPPRESSOR GENE, IS EXPRESSED IN HUMAN BASAL BREAST EPITHELIAL CELLS BUT NOT IN BREAST CARCINOMA CELLS By MakiSaitoh ATHESIS Submittedto MidnTganStateUniversity inparthlfulfillmentoftherequirements forthedegreeof MASTEROFSCIENCE GemtimProgram 2004 ABSTRACT MASPIN, A TUMOR SUPPRESSOR GENE, IS EXPRESSED IN HUMAN BASAL BREAST EPITHELIAL CELLS BUT NOT IN BREAST EPITHELIAL STEM CELLS AND BREAST CARCINOMA CELLS By Maki Saitoh The phenotypes of cancer cells are very similar to stem cells as compared to differentiated somatic cells. Both are described as undifferentiated cells and share the common expression of many genes related to tumorigenesis, a basis for the concept that cancer cells are primarily derived from stem cells. Maspin, a protease inhibitor known to regulate not only invasion and metastasis, but also tumor growth and apoptosis, could be a tumor suppressor gene not expressed in stem cells and tumor cells. To test this hypothesis, I have examined the expression of maspin, using immunocytochemical and western blot techniques, in two types of normal human breast epithelial cells (HBEC), Type I HBEC show stem cell characteristics, whereas Type II HBEC expressed basal epithelial cell phenotypes. The results clearly show that Type I HBECs, indeed, did not express maspin in contrast to Type II HBECs which highly expressed the maspin. A series of Type I HBEC lines, neoplastically transformed at different stages (immortal, weakly and highly tumorigenic) and three breast carcinoma cell lines were also not found to express maspin. Thus, the silencing of maspin expression in breast cancer cells is very likely due to the continuous non-expression of a target stem cell phenotype. The results provide additional evidence for the stem cell theory of carcinogenesis and indicate that maspin could be used as a marker for early detection of breast cancer and for assessing the efficacy of chemopreventive or chemotherapeutic agents. ACKNOWLEDGEMENTS hismypleasmemaiqnwlodgeflnnmypeoplewlnmsistedmflwppmedmeovaflw omnseofmygradmtework. MydeepestfliarflcsgnomymajoradvisonDLGfia-Clmgflmgwho gidedmflmgtaflaagesofmymepogmnmmspofomflhfigmammsdmomdm andwarmsupport. Ifisbdiefmmeamnagedmetokeepwoddngmdlfisdevotedmdflmghtfifl attituletowardmeasaneduoatortaughtmegreataspeotsoflife. MysimaegafiuxlegmmDrstEdecowlmofl‘aedmegwamwaodlan suggestions. Imndoeplyappredafiveoftfisvalmbleadvioafimeandmpport BmmsdmlsmersBradJUplmed-HtfiTaiTsai-MmngmgKng-I GmsKymg-SmesWd SmGfing-“HsidLAngelaCdeindaWflsmeKymgNa, GmgOmImjiSmfliNWDeoCmnmfioflemDowfingforflrdrflmglfihLommxfiveadWoe germssupportandgaminefiiendship. Timflcsformyfellowgradmtemrtsandtednficians, Arrthatoh,An-Pde,Kai—IhmnggarxlEm—HymAm Theybelievedinmebeforel bdievedmmysdflwfllnevahaveanighwmdsmmlnwinpmtmntheymemme iii TABLE OF CONTENTS List ofTables and Figures Literamre Review L'lheRoleofStanCdlinCarcinogenesis ILCanoerCellthotypes 1)Ijmitlelsreplimtivepotential 2) Self-sufficiency in growth signals 3) Insensitivityto growth-inh‘bitmy signals 4)Evasionofprogrmnnedoelldeath S)Sustainedangiogams @Tmmirwasimandmetaaas's I. GmesAlta'odinBieastCamer 00mm 2)“!‘umorSuppmsorGenes IL Maspin 1) Identification and location. 2) Fundim 3) Transcriptional Regilation Irmotmdim MataialsarxiMethods CellCulture 1)NonnalenanBreaaEpithelialCefls 2)TmmigenicBreastEpiflieIialCells WestanBlaAnalysisofMaqrinProteinExpression hmnmncytodremimlDetedimofMawinEmmsim Results DiffuafialExprafionofMaspininTypeIandTypelIHBEC EmessimofMaspinIanoTransfmnedHBECarxlCaerells D' . RefamoeList iv \OOOOQQ-b-b—‘I-‘s. 38318883881?!ng LIST OF TABLES Table ICIIaIaaaistiqsonypeIaerTypeIII-IBECS 77 LISTOF FIGURES FigmellsolafimsdmiemobtainZtypesomenalemBreastEpiflidialCdls .................. 23 Figure 2 In vitro immortalization and neoplastic transformation of Type I HBEC...25 Figme3MuflnlogiesonypeIaerTypelII—IBECS 79 Pigne4ExprmsimCytokeratinl4andl8inTypeIarflTypeHI-IBECS 30 Figme SaMmrinExpmsionbyhmnmostairfinginTypeIGefi)andTypeII (right) HBEC ofHME 30 32 FigueSbMasrfinExpmssimbyhmnmostairfinngypeIGefieriTypeH(right)I-IBECole\rIE 31 33 Figme6WrstanBlotAmlysisofM$pinExpresioninTypeIamiTypeIII—IBECofWGanes 1,2),I-ME300m3,4)andI-IME31(Im5,6) 34 Figure7MasphiEiqxesimoflnviuoTrmsfimnedTypeIHBECIjnesatDifi‘aanSmby hmnmocytochanioalAnalysis 36 Figme8MmpinE>qxessiminBreastCardnmnaCdlLinesbyhmnmnstainh1g 37 Figm9Westm1BIdAmly§sofMaq3hE7qxessimmNommlHBECmvimlhmnmmhzodm NeoplasfimflyTramfixmodTypeIHBECandBreastCm'cirnnaCdlfirm 38 vi LITERATURE REVIEW I. The Role of Stem Cell in Carcinogenesis Several lines of evidence suggests that cancer cells are derived from the adult stem cell 4 pools of a tissue where initiating events of stem cells prevents cells from fully differentiating and resulted in uncontrolled clonal expansion of these cells. The stem cell theory of carcinogenesis proposes that stem cells are target cells in carcinogenesis and certain developmental or differentiating genes are blocked or partially blocked so the cells remain undifferentiated (Potter, 1978). Stem cell research has received increasing attention because of its therapeutic application and potential role as a target cell in carcinogenesis. Its unlimited or extensive self- renewal capacity and ability to differentiate into multiple cell types (“pluri-potency”) are highly valuable in various applications for medical treatments such as tissue engineering, transplantation and gene therapy. These two unique properties of stem cell are also important in understanding the role of stem cell in carcinogenesis. By definition, "toti-potent" cells can give rise to all cell types within the organism. They can divide symmetrically or asymmetrically to produce partially committed organ specific stem cells, namely "pluri-potent" stem cells. These pluri-potent stem cells can give rise to several different cell types in different organs. Then, after further development, some cells are finally restricted to giving rise to one or fewer cell types to become "committed" progenitor cells. The committed progenitor cells eventually develop and give rise to functionally and terminally differentiated cells. The stem cell theory of carcinogenesis states that the carcinogenic process starts from a stem cell (most likely a pluri-potent stem cell), assuming that a normal stem cell is "immorta " (Trosko, 2003) As an alternative to the stem cell theory, the de—differentiation theory states that cancer cells are derived from de-differentiation of differentiated of cells to explain the undifferentiated state of tumor cells. The unlikelihood of dedifferentiation of differentiated somatic cells to develop into tumor favors the stem cell theory. For example, many differentiated hepatocytes, are tetraploid (Styles et al., 1985), while the nodule that give rise to tumors are diploid (Schwarze et al., 1984). It is a rare chance, therefore, that these cells can de-differentiate back to diploid state that characterize precancerous cells and this fact seriously undermines the de-differentiation theory as a possible explanation of less differentiated state of cancer cells. It has been documented that the stem cells share a number of tumor cell characteristics: contact insensitivity (Chang et al., 1987), extensive self-renewal capacity (Tang et al., 2001; Mathon et al., 2001), anchorage- independent growth (Chang et al., 2001; Chang et al., 2004; Lin et al., 2004), lack of gap junctional intercellular communication (6] 1C), (Y amasaki et al., 1987; Chang et al., 1987; Kao et al., 1995; Matic et al., 2002; Grueterich and Tseng, 2002) and activated telomerase (Hiyama et al., 1995) or high susceptibility of telomerase activation (Sun et al., 1999) . This evidence also indicates the difficulty to de-differentiate back to undifferentiated state to regain those characteristics described. Recently, more evidence suggests the role of stem cells as target cells in carcinogenesis. For example, McKay and his co-workers found a novel protein named nucleostemin, from a subtractive hybridization study, that was expressed in the nucleoli of CNS stem cells and embryonic stem cells, as well as in tumorigenic cells (Tsai and McKay, 2002). Structural analysis and mutation study of nucleostemin indicated a role of the gene in cell cycle regulation of cell growth, as well as the prevention of the cells from dying by apoptosis. The expression of nucleostemin decreased rapidly prior to cell cycle exit when the cells are differentiated. Furthermore, based on the evidence that activation or over- expression of nucleostemin reduces cell proliferation in CNS stem cells and transformed cells, nucleostemin seems to be correlated with cell proliferation or the lack of differentiation in these cells. Recently, the role of myoepithelial cells as a host defense cell against cancer received some attention (Sternlicht and Barsky, 1997; Stemlicht et al., 1997). First, the mammary myoepithelial cells express protease inhibitors, including maspin, which have various tumor suppressive activities. Second, myoepithelial cells are rarely transformed and the myoepithelial tumor tends to be benign or low-grade neoplasms. Third, the myoepithelial cells promote differentiation and inhibit invasion in vitro. In terms of the stem theory of carcinogenesis, the properties of myoepithelial cell show that they are differentiated cells derived from stem cells and that they possess anti-cancer properties. The fact suggests that the difierentiated breast epithelial cell type is unlikely to be a target cell and indirectly supports the stem cell theory of carcinogenesis. Evidence supporting the stem cell theory of carcinogenesis has been previously reviewed (Chang et al., 2001) and mentioned in the Introduction. 11. Cancer Cell Phenotype Hanahan and Weinberg summarized six essential capabilities of cancer cell or "hallmarks of cancer": 1) limitless replicative potential, 2) self-sufficiency in growth signals, 3) insensitivity to growth-inhibitory signals, 4) evasion of programmed cell death, 5) sustained angiogenesis, and 6) tissue invasion and metastasis (Hanahan and Weinberg, 2000). Now, in view of the stem cell theory, I will briefly summarize them to re- emphasize how the stem cell already possesses several important characteristics commonly found in the tumor cell. 1) Limitless replicative potential Normal human cells have limited life spans (Hayflick, 1997), however, almost all malignant tumor cells can replicate indefinitely. In view of the stem cell theory, stem cells theoretically have extensive self-renewing capacity. More specifically, the stem cells or their partially differentiated daughter cells, that have not yet down-regulated their telomerase activity, but are exposed to a carcinogenic "initiator," will remain immortal or have limitless replication potential (Trosko et al., 2000). Thus, the first step of carcinogenesis is to prevent the stem cells to become mortal or to terminally differentiate. Although the stem cell has extensive self-renewal capacity, it is not all clear whether stem cell maintains this capacity for an entire lifespan and if so, what type of intrinsic or extrinsic factors are involved to maintain this capacity. Several in vitro studies indicate that certain genetic alterations are required to maintain the self-renewal capacity. The induction of so-called "immortalization" of human mammary epithelial cell (HMEC) seems to involve two major events (Kiyono et al., 1998). First, the cell has to acquire an extended lifespan (to overcome that first stage senescence or mortality stage, M1) by altering the regulation of cell cycle genes especially the p16ink4/Rb pathway (Kiyono et al., 1998). Inactivating this p16ink4/Rb pathway to extend the lifespan of HMEC has been demonstrated by transfecting with SV40 large T-antigen (Kao et al., 1995) or papilloma virus E6 and E7 (W azer et al., 1995). However, the cells that successfully evaded the senescence soon stop growing when they reach the next plateau, called crisis (Kiyono et al., 1998). As a second step in immortalization, the HBEC with extended lifespan may become immortal after the activation of telomerase (to overcome the M2, or crisis). HMEC with inactive p16/Rb may become immortal after transfecting with the human telomerase gene hTERT (Kiyono et al., 1998). However, HMECs, growth on feeder layers, have been immortalized without abrogating p16 (Herbert et al., 2002). It is interesting to note that estrogen is an agent known to activate telomerase on the hTERT promoter in a breast cancer cell line (Kyo et al., 1999). The capability of unlimited or prolonged replicative potential is believed to be established in early stage of carcinogenesis. This may be related to the characteristics of stem cells, the major target cells for carcinogenesis. The unlimited self-renewal ability of stern/precursor cells has been demonstrated for rodent cells (Tang et al., 2001; Mathon et al., 2001) but not for human cells. However, breast epithelial stem cells have been shown to be more susceptible to telomerase activation (Sun et al., 1999). Taken together, to preserve the limitless self-renewal capacity of stem cells, alteration of cell cycle gene regulation and activation of telomerase gene are required to occur during the development and growth of cancer 2) Self-sufficiency in growth signals Normal cells require mitogenic growth factors, whereas tumor cells produce and express many of their own growth factors and growth factor receptors, thereby reducing their dependence on exogenous or endogenous growth factors fi'om their normal tissue microenvironment. The most important growth factor for breast cancer cells appears to be the estrogen. In breast cancer patients, about two-thirds of tumors are ER-positive (Lippman et al., 1988). Fifty percent of these ER-positive tumors are estrogen-dependent and respond to endocrine therapy and anti-estrogen, Tamoxifen (Manni et al., 1980). The estrogen induction of proliferation of breast cancer required the expression of Myc (Watson et al., 1991) which is frequently amplified or overexpressed (Dickson, 1994). The over- expression of the neu oncogene might enable the cancer cell to become hyper- responsive to ambient level of growth factors that normally would not trigger proliferation in normal cells (Fedi, 1997). In fact, amplification of the EGF-receptor- related prooncogene c-erbB2/neu has been observed in 30% of primary human breast carcinomas (Slamon et al., 1987) and another 10% over-expressed c-erbB2 without amplification of the gene (Kraus et al., 1987). It should be noted that, in contrast to breast epithelial cells, human breast epithelial stem cells express the estrogen receptors (Kang et al., 1997). 3) Insensitivity to growth-inhibitory signals In normal cells, a number of anti-proliferative signals, including grth inhibitors (Hanahan and Weinberg, 2000), operate to maintain cellular quiescence and tissue homeostasis. However, cancer cells have an ability to evade these anti-proliferation signals and use various strategies to avoid being terminally differentiated. As an anti- growth factor, TGF-beta, prevents the phosphorylation of Rb, subsequently blocks the cell to advance through the GI phase of the cell cycle (Hanahan and Weinberg, 2000). Endogenous TGF [3 signaling and growth suppression is lost during human mammary epithelial cell transformation by benzo(a) pyrene (Ge and Stanpfer, 2004). Down- regulation of GJIC is one of the mechanisms to prevent fiom being regulated by growth inhibitory signals, such as cyclic AMP, which is considered as a reverse transformation agent (Chan et al., 1989). It has been reported that Type I HBEC with stem cell characteristics and most breast carcinomas are deficient in GJIC (Kao et al., 1995). Another mechanism to avoid being terminally differentiated is the over-expression of the survival factor c-myc, which encodes a transcription factor. During the normal development, the grth stimulating action of Myc is associated with another factor, Max. The association can be supplanted by alternative complex of Max with a group of Mad transcription factors; the Mad-Max complexes elicit differentiation-inducing signals (Foley and Eisenman, 1999). Overexpression of c-myc can reverse this process, shifting the balance back to favor Myc-Max complex, thereby impairing differentiation and promoting growth (Hanahan and Weinberg, 2000). In breast cancer cells, the abnormalities in the c-myc gene were reported in about 6-32% of breast cancer (Van de Vijver and Nusse, 1991 ). TGF-beta also suppresses the expression of the c-myc gene, which regulated the 61 cell cycle (Moses et al., 1990). ER-positive breast cancer cells are refractory to TGF-beta effects, whereas ER—negative breast cancer cells are often TGF- beta sensitive (Arteaga and Moses, 1996). 4) Evasion of programmed cell death The balance between cell proliferation rate and cell attrition rate also determines the ability of tumor cell populations to expand in number. Apoptosis or programmed cell death is an active, energy-dependent process of cell death which occurs during development, in response to certain physiologic stimuli (Kerr et al., 1972). Unlike necrosis which is a passive osmosis-driven process due to the loss of ion-pumping activity, apoptosis shows characteristic morphologic changes i.e. cell shrinking and blebbing, nuclear and chromatin condensation, nuclear fragmentation, and packaging of cellular materials and organelles into mernbrane-bound vesicles termed apoptotic bodies (Hahm, 1998). Once cells are triggered by a variety of physiologic signals, the cells undergo apoptosis in a precisely choreographed fashion. There are two major protein families involved in apoptosis: antiapoptotic protein family (Bel-2 relatives) and proapoptotic protein family (Bax relatives) during induction stage of apoptosis. The tumor suppressor protein p53 can elicit apoptosis by upregulating the expression of Bax in response to sensing DNA damage. 5) Sustained angiogenesis Angiogenesis, the growth of new capillary blood vessels, is critical for cancer cell to grow, invade and metastasize. New blood vessels bring in fresh nutrients and grth factors so that the tumor mass can expand. As a matter of fact, without forming new blood vessels, solid tumors cannot exceed a size of about 1mm3 . Interaction between angiogenic proteins (i.e. VEGF, FGFs) and angiogenic proteins (i.e. thrombospondin) appear to be responsible for the angiogenic activity of malignant tumors. Interestingly, analyses of histologically distinct stages of human and rodent tumors suggest that the acquisition of an angiogenic phenotype occurs early in tumorigenesis and can be considered the rate limiting step during tumor progression (Hanahan and Folkman, 1996). 6) Tissue invasion and metastasis In order for cancer cells to spread, the cells need to acquire the ability to invade the tissue and enter the blood stream to metastasize to other organs. These involve cell migration and the expression of proteases. It has been shown that increased expression of hepatocytes growth factor (HGF) and its receptor, met, occurs in invasive human breast cancer (Tuck et al., 1996; Wang et al., 1994; J in, 1996) particularly at the migrating tumor fi'ont (Tuck et al., 1996). The tissue invasion and metastasis involve the action of adhesion molecules, metalloproteases and cllageneses (Akiyarna et al., 1996). III. Genes Altered in Breast Cancer As stated by Hanahan and Weinberg (2000), “tumors are the results of the accumulation of a series of genetic alterations. Genetic studies over the years have identified a number of oncogenes and tumor suppressor genes associated with the development of breast cancer (See ref. In Chang et al., 2001). i) Oncogenes Three oncogenes (c-erb BZ/neu, c-myc and int-2) and the G. phase cyclins (D and E) were frequently found to be amplified or overexpressed in breast cancers (See ref. In Chang et al., 2001). In addition, many breast oncogenes have been identified. These include Brk (breast tumor kinase) (Barker et al., 1997; Mitchell et al., 1994), srk (Jacobs and Rubsamen, 1983; Luttrell et al., 1994), AKT3 (Nakatani et al., 1999) and ZN F217 (Nonet et al., 2001). ii) Tumor Suppressor genes 10 As summarized (Chang et al., 2001 ), many tumor suppressor genes are involved in breast cancer including those responsible for breast cancer syndromes (i.e. BRCAl, BECA2, p53 in Li-Fraumeni syndrome, ATM gene in ataxia telangiectasia). In addition, PTEN (a lipid phosphatase), P57klpz (a cyclin kinase inhibitor), F HIT (fragile histidine triad), Wnt- 5a, pl6ink4a (a cyclin-dependent kinase inhibitor) and maspin are also likely to be involved in breast cancers. Loss of heterozygosity was frequently observed on many chromosome arms in breast cancer (i.e. 1p, lq, 3p, 11p, 13q, 16q, 17p, 17q and l8q). Except for chromosome 1, 16q and 18q, potential turner suppressor genes have been identified (Chang et al., 2001). Subsequently, a senescence gene, SEN16 (Reddy et al., 1999; Reddy et al., 2000)and a transcription repressor, CBFA2T3 (Kochetkova et al., 2002) have been located on 16q24 and the maspin gene has been mapped to 18q21.3 (Schneider et al., 1995). IV. Maspin The mammary serpin protease inhibitor, maspin, is a candidate for being a tumor suppressor gene whose expression is lost in many advanced breast tumors. Maspin has drawn the attention due to the various proposed functions in breast carcinogenesis, such as angiogenesis, metastasis, apoptosis and cell invasion. 1) Identification and location The maspin gene was originally isolated from normal mammary myoepithelial cells by subtractive hybridization on the basis of its expression at the m-RNA level (Sager et al., 11 1994). This maspin gene encodes a 42-kDa protein (Sager et al., 1994) and is mapped at 18q21.3 (Schneider et al., 1995) where the loss of heterozygosity is frequently observed in this region , not only breast cancer cells, but in a large number of human tumors (Schneider et al., 1995). Maspin m-RNA and protein were found in normal mammary myoepithelial cells, but down-regulated in mammary carcinoma cells (Sager et al., 1994). The expression of maspin is tissue-type and cell-type specific and it is expressed in skin keratinocytes and prostate epithelial cells in addition to mammary epithelial cells (Futscher et al., 2002). Immunohistochemical studies showed that maspin is expressed in pancreatic cancer cells but not in normal pancreatic epithelium (Maass et al., 2001a). In contrast, in mammary tissue, maspin is expressed in normal epithelial cells, but significantly less in mammary tumor cells (Maass et al., 2001a). In contrast to p53, the maspin expression was higher in ER-negative tissue than ER-positive tissue (Martin et al., 2000). Based on immunohistochemical studies, the maspin is expressed in the nucleus, as well as in the cytoplasm of breast myoepithelial cells (Reis-Filho et al., 2001; Lele et al., 2000). 2) Function Although it has not been clearly understood the role of maspin in breast cancer, there are many functions of maspin suggested since the discovery of the maspin gene in 1994. Unlike the other serpins, maspin does not act as an inhibitor to an array of serine proteases, including urokinase-type plasminogen activator (uPA) and tissue-type plasminogen activator (tPA) (Martin et al., 2000; Pemberton et al., 1995). However, 12 there is evidence that tPA is a target of the tumor suppressor gene maspin (Sheng et al., 1998). Exogenously introduced maspin gene expression in breast tumors rendered the inhibition of cell invasion in vitro and metastasis in vivo (Sager et al., 1994). Recombinant maspin protein blocks the motility of carcinoma cells (Sheng et al., 1996). Neutralization of maspin by an anti-maspin antibody abolished the invasion suppressive effect and increased apoptosis of conditioned medium from cultured breast myoepithelial cells on tumor cells (Shao et al., 1998). Maspin overexpression increased a rate of apoptosis and block angiogenesis in vivo and in vitro (J iang et al., 2002; Zhang et al., 2000a; Zhang et al., 2000b). Maspin expression in breast tumor cells reduces tumor induction and metastasis in nude mice (Sager et al., 1994; Shi et al., 2001; Shi et al., 2003; Streuli, 2002), and also invasion of the basement membrane in vitro (Sager et al., 1994). Moreover, treatment of human breast cancer cells with recombinant maspin (rMaspin) inhibits cell motility (Sheng et al., 1996). 3) Transcriptional Regulation The molecular and biological mechanisms of the functions of maspin still remain unknown. However, there is evidence that maspin interacts with the p53 tumor suppressor pathway (Maass et al., 2000). Zhou reported that p53 regulates the expression of maspin in breast cancer cell lines (Zou et al., 2000). Maspin expression was up- regulated when wild-type p53 gene was introduced through an adenoviral vector. P53 activates the maspin promoter by binding directly to the p53 consensus-binding site present in the maspin promoter (Zou et al., 2000). Furthermore, they demonstrated that 13 DNA-damaging agents and cytotoxic drugs induced endogeneous maspin expression in cancer cells containing wild-type p53, but not in cells containing mutant p53 (Zou et al., 2000). These results indicate that the function of maspin in apoptosis might be regulated through the p53 pathway. Although the maspin gene is silenced in cancer cells, maspin gene deletions and mutations have not been found (Barsky et al., 1997), indicating the involvement of an epigenetic mechanism. The mechanism of the maspin gene regulation has not been clearly understood. However, maspin is up-regulated by the treatment of gamma linoleic acid, an essential fatty acid with anticancer properties (Jiang et al., 2002). Maspin reacts with tissue type plasminogen activator in vivo (Sheng et al., 1998). Interestingly, recombinant maspin exerts a biphasic effect on the activity of single-chain tissue plasminogen activator, acting as a competitive inhibitor at low concentrations (<0.5 mM) and as a simulator at higher concentrations (Sheng et al., 1998). Maspin gene expression was induced by MnSOD (Manganese-containing superoxide dismutase cDNA) transfectants of MCF-7. (Li et al., 1998). The involvement of DNA methylation was first suggested by Domann in 2000 (Domann et al., 2000). Maspin promoter is unmethylated and is an open chromatin structure in normal, maspin-positive HMECs. In contrast, this chromatin structure is often aberrantly methylated and associated with a closed chromatin structure in maspin-negative human breast cancer cell lines (Domann et al., 2000). Methylation-associated silencing is found with a high frequency, as high as 80% in human breast cancers (Domann et al., 2000). 14 Additional experiments confirmed that maspin expression is regulated by DNA methylation and/or histone deacetylation in maspin-negative breast cancer cell lines (Domann et al., 2000; Futscher et al., 2002; Mass et al., 2002). 15 INTRODUCTION Cancer cells are generally known to be similar to stem cells in phenotypes. The extreme case is exemplified by human embryonic stem cells (ES) which formed teratomas in SCID-beige mice (Thomson et al., 1998). Of all the common characteristics, the most notable description is the undifferentiated state of cancer and stem cells. The undifferentiated state of cancer cells could be due to the de-differentiation of differentiated somatic cells or due to the blocked differentiation of stem cells which give rise to tumor cells (Varmus and Weinberg, 1993; Sell, 1993). The latter is similar to other concepts of cancer as a disease of cell differentiation (Markert, 1968), a disease of stem cells (Sawyers et al., 1991) or as an “oncogeny as blocked or partially blocked ontogeny” (Potter, 1978). Cancer cells are known to be deficient in homologous or heterologous junctional gap intercellular communication (GJIC) (Y amasaki et al., 1987). Several human adult stem cells were also found to be deficient in GJIC (Chang et al., 1987; Kao et al., 1995; Matic et al., 2002; Grueterich and Tseng, 2002; Chang et al., 2004). Some characteristic tumor cell phenotypes such as contact-insensitive growth and the ability of anchorage independent growth (AIG) are shared by stem cells. The former has been reported for human kidney epithelial stem cells (Chang et al., 1987) and the latter were shown in human breast epithelial, human liver and mesenchymal stem cells (Chang et al., 2001; Chang et al., 2004; Lin et al., 2004). Nucleosternin, a nucleolar protein that interacts with p53, is present in CNS stem cells, ES cells and cancer cells but not in the differentiated cells of adult tissues (Tsai and McKay, 2002). Similarly, Oct-4, a transcription factor previously reported to be exclusively expressed in pluripotent early 16 embryo cells, ES cells and undifferentiated tumor cells, has been recently shown to be expressed in many human adult stem cells (Trosko et al., 2004). Furthermore, the expression of ot-fetoprotein and vimentin, collectively termed the “oval cell phenotypes” (Alison et al., 1997), was found in adult human liver stem cells and hepatomas (Chang et al., 2004). Mammary cancers primarily originated from the relatively undifferentiated mammary gland, i.e. the rodent terminal end bud (TEB) or the human lobule 1 where most mammary epithelial stem cells reside. For example, it has been demonstrated that the carcinogen acts on TEB and that the structure is the one that evolves to intraductal proliferation, carcinoma in situ and invasive carcinoma (Russo and Russo, 1996). The study of pathogenesis of breast cancer in relation to the lobular composition of breast also identified lobule 1 as the site of origin of the most frequent breast malignancy, the ductal carcinoma (Russo and Russo, 1997). Consistent with this correlation is the observation that lobule 1 contains high frequency of estrogen-receptor (ER) positive cells (14%) compared to lobule 2 (4%) and lobule 3 (0.5%) (Russo and Russo, 1998), and the fact that two thirds of breast cancer is ER-positive (Lippman and Allegra, 1980). At the cellular level, the mammary gland contains two types of epithelial cells, the lurninal and the basal (or myoepithelial in alveoli) epithelial cells besides the epithelial stem cells. Our laboratory has developed a cell culture method to grow two types of human breast epithelial cells (HBEC) from reduction marnmoplasty tissues (Kao et al., 1995). Type II HBEC expressed basal epithelial cell markers whereas Type I HBEC l7 showed luminal epithelial and stem cell phenotypes. The stem cell characters of Type 1 cells include 1) deficiency in GJIC; 2) the ability to differentiate into Type II HBEC; 3) the expression of Oct-4 (Trosko et al., 2004) and 4) the ability to form ductal and budding or lobule l-like structures on Matrigel (Chang et al., 2001). Furthermore, Type I HBEC have been shown to be more susceptible to telomerase activation, immortalization and neoplastic transformation (Sun et al., 1999; Kang et al., 1998). When the phenotypes of breast carcinomas or in vitro neoplastically transformed Type I HBEC are compared with Type I and Type II HBEC, it is clear that tumorigenic breast cells are similar to Type I HBEC rather than Type II HBEC. These similarities include deficiency in GI 1C, the expression of estrogen receptor, Oct-4 and luminal epithelial cell markers (i.e. epithelial membrane antigen, cytokeratin 18) and ability of AIG (Kao et al., 1995; Kang et al., 1997; Chang et al., 2001; Trosko et al., 2004). Since stem cells and differentiated cells are substantially different in phenotypes as shown in Type I and Type II HBEC, and if cancer cells are derived fi‘om and preserve many stem cell characteristics, it is expected that the phenotypes of the early stage cancer cells are already very different fiom differentiated normal cells. Indeed, it was found that the most dramatic and consistent phenotypic change occurred at the normal-to-in situ carcinoma transition (Porter et al., 2001). Another implication is the doubt about genetic alterations in cancer cells based on differential display or microarray studies of cancer and normal cells since the great majority of the cells in normal tissues are not the stem cells from which most cancers arise. For example, Cx26 and a6 integrin and maspin have been considered as tumor suppression genes based on differential display studies 18 showing their expression in normal cells but not in tumor cells (Lee et al., 1991; Sager et al., 1993; Zou et al., 1994). In fact, our results show that Cx26 and a6 integrin were expressed in Type II HBEC but not in Type I cells (Kao et al., 1995; Chang et al., 2001). Bemards and Weinberg (Bemards and Weinberg, 2002) hypothesized that certain combination of genetic alterations that are selected relatively early in tumorigenesis for proliferation advantage they confer will, incidentally, also confer invasive/metastatic phenotype. Since maspin is known to be a protease inhibitor which regulates not only invasion and metastasis but also tumor growth, angiogenesis and apoptosis (Zhang et al., 2000b; Jiang et al., 2002; Streuli, 2002; Shi et al., 2003), it is hypothesized that this gene could be the kind of tumor suppressor gene not expressed in stem cells and tumor cells. The experiments of this study were carried out to investigate this hypothesis. l9 MATERIALS and METHODS Cell Culture a) Normal Human Breast Epithelial Cells Breast tissues were obtained from healthy women who underwent reduction mammoplasty. After removing most adipose tissues, the tissues were minced into small pieces and digested with collagenase for overnight before culturing in the MSU-l medium (Kao et al., 1995). The initial primary culture developed in one week were aliquoated in small vials and stored at —80°C liquid nitrogen storage until use. To recover cells from liquid nitrogen storage, the cells were thawed at 37°C and transferred to a 15 ml centrifuge tube containing MSU-1 medium. The tube was then centrifuged at 1000 rpm for 10 min. After removing the freezing solution and the medium fiom the tube, the cell pellet was plated in 6 ml of MSU-I medium with 5% fetal bovine serum (PBS) (the Type I medium) in a 10 cm cell culture dish (Plate A) and incubated for two hours. After the incubation, the supernatant containing unattached cells was removed, transferred to a 15 ml centrifuge tube and centrifuged at 1000 rpm for 10 min. The Plate A containing Type I HBEC and fibroblast cells was further incubated after adding 6 ml of fresh MSU-I medium. The cell pellet was suspended in 6 ml of MSU-I medium with bovine pituitary extract (BPE) (50 til/ml) (the Type 11 medium), plated in a 10 cm cell culture dish (Plate B) and incubated for overnight. Next day, the supernatant containing unattached cells was carefully removed and transferred to a 15 m1 centrifuge tube and centrifuge at 1000 rpm for 10 min. After discarding the medium, the 20 cell pellet was suspended in 6 ml of MSU-I medium with 5% FBS and plated in a 10 cm dish (Plate C), which contains mostly Type I HBEC and few fibroblasts. The few colonies of fibroblasts that developed in a week can be marked and removed by scraping with a rubber policeman. The Plate B after further incubation in the Type 11 medium contains Type II HBEC. The procedure of obtaining these 2 types of HBEC is diagrammatically depicted in Fig. 1. For this study, the primary HBEC cultures HME 29, 30 and 31 derived from 3 different women (age 26, 23, 21 respectively) were used. The phenotypic difference between Type I and Type II HBEC fi'om previous studies (Kao et al., 1995; Chang et al., 2001) are listed in Table l. The culture medium was changed once every three days. Subculture of HBEC was carried out by cell removal with 0.01% trypsin and 0.01% EDTA followed by the inactivation of trypsin with 10% PBS. 21 Table 1 Characteristics of Type I cells and Type II HBEC (Modified from (Kao et al., 1995) Plastic Type I Type II Cell Morphology in Variable in shape Uniform in shape, cobble- MSU-l Medium stone appearance Colony Morphology in Boundary smooth and Boundary not smooth, MSU-1 Medium restricted unrestricted Attachment on Plastic After Late Early Epsinization Effect of FBS on Cell Growth Promoted Inhibited EMA Expression + - Keratin 18 Expression + — Keratin 14 Expression - + a6 Integrin Expression - + Connexin 43 ~ + Connexin 26 - + Effect of Cholera Toxin Induction of Type I into Growth promotion Type II HBEC AIG on Soft Agar + - Telomerase Activation and High Frequency Low Frequency Irnmortalization by SV-40 Large T Antigen Organoids Structures Formed Acini Hollow Spheres on Matrigel Budding/ductal structure in conjunction with Type II HBEC Gap Junctional Intercellular Deficient Proficient Communication 22 Storage of HBEC, developed in MSU-l medium for 1 week, in liquid nitrogen Thawi g HBEC 2 hours . Centrifugation of Supernatant 111911939011 in Growth of Pelleted Cells Type I Medium Centrifugation of Atta h ed Overnight Incubation in Supernatant ecu:- Type 11 Medium Growth of Pelleted Cells HBEC and ‘ fibroblasts \\\\\\\\\ Attached Cells: Type II HBEC Attached Cells: Type I HBEC Figure 1. Isolation scheme to obtain 2 types of Normal Human Breast Epithelial Cells 23 b) Tumorigenic Breast Epithelial Cells Immortal, weakly tumorigenic and highly tumorigenic breast epithelial cell lines have been derived from Type I HBEC after step-wise treatment with SV40 large T-antigen, X- rays and c—erbB2/neu oncogene (Kang et al., 1998) (Fig. 2). These in vitro derived cell lines together with the well known breast carcinoma cell lines, MCF-7 (ER-positive) and MDA-MB-231 (ER-negative), were also used in this study. 24 HME 13 Type I HBEC Transfection with SV40 Large T-antigen V M13SV1 (Immortal) X-ray irradiation Selection of fast-growing colonies M13SV1R2 (Weakly Tumorigenic) Transfection with a mutated neu oncogene ‘M13SV1R2N1 (Highly Tumorigenic) Figures 2 In vitro immortalization and neoplastic transformation of Type I HBEC (Kang et al., 1998) 25 Western Blot Analysis of Maspin Protein Expression Cells cultured in 10 cm dish, were washed with phosphate buffered saline (PBS) three times, and lysed with 20% SDS lysis solution (500 pl) containing protease and phosphatase inhibitors (Trosko et al., 2000). The viscous lysate was carefully collected with a scraper and transferred to a tube on ice. The lysate in the tube was sonicated with a sonicator (range~33) for 30 seconds 3 times. The proteins were aliquated in smaller tubes and stored in -20°C freezer until use. Protein concentrations were measured using Biorad Protein Quantification kit (Biorad, California). Equal amount of protein (20 ug/lane) were separated by 12% SDS-PAGE and transferred from the gel to PVDF membranes (Millipore Corp, Bedford, MA). Irnmunoblotting was carried out using anti-hmnan maspin antibodies (BD Biosciences- PharMingen, San Diego, CA, 1:1,000 dilution). Horseradish peroxidase-conjugated secondary antibody was used for the chemiluminescent assay. The immunoreactive protein complexes were detected by ECL-detection kit (Amersham, Life Science, Denver, CO). When the membrane was needed to be reblotted, the membrane was washed in 100 ml of 0.1 M citrate, pH=3.5, for 10 minutes. Then, the membrane was further washed in 100 ml Tris-buffered Saline (TBS) containing 0.5 M tris (pH=7.5), 0.05M NaCl and NP40 (IGEPAL) (0.5% v/v) for 20 minutes. After repeating the above two steps twice, the membrane was washed twice with 100 ml TBS without IGEPAL for 10 minutes. After 26 this step, the membrane was reblocked with 5% milk in PBS with 0.1% Tween before initiating the re-probing. Immunocytochemical detection of maspin expression Cells were plated into 8-well chamber slide (LAB-TEK, Nalge Nunc Int., Naperville, IL; 12,000 cells/well) for 2-3 days. The cells were rinsed with PBS and fixed with 4% paraformaldehyde in PBS for 30 min. After rinse with PBS, the cells were permealized with 3% H202 in methanol at room temperature for 10 min and treated with 10% FBS in PBS for 30 min for blocking. These cells were then treated with primary antibody (a mouse antihuman maspin monoclonal antibody from BD Biosciences-Pharmingen, Cat. #554292, 1:100 dilution) for overnight at 4°C on a rocking platform, following rinse with PBS, the cells were incubated with the secondary antibody (i.e. FITC-conjugated sheep anti-mouse IgG, 1:200 dilution in 1% FBS in PBS) for 45 min at 4°C. The cells were thoroughly washed with PBS and mounted with aqua-poly mount (Polysciences Inc. Cat#18606) and a cover glass. The image and fluorescence of cells were observed and recorded using a Nikon Eclipse TB 300 microscope connected to a digital camera and computer. Images in this thesis are presented in color. 27 RESULTS Difl'erent'mlexpressionofmaspininTypeIand'I‘ypeIIHBEC TypeImflTypeHHBECswaeobminedfimreducfimnmmplaayfimudngmesdrane describedinFig 1 bytaldngadvantageofflredifi‘amcemfimereqtnredforceflanadnnaumnoflre plasticdishesaftercellplating Fmflmnmeflresehvotypesofl—IBECmnbeselectivelygrownusing difi‘aartmediaieTypeIMSU-l medimnwiflrFBSpromotedthegrowthonypeII-IBEthile inhibitedthegrowflr onypeII HBEC;TypeII MSU-1 mediurnwith BPEpromotedthegrowth of TypeIIHBECandtheconversiononypeItoTypeIII-IBEC. TypeIandTypeHHBECrfisplayawholesalerfifi‘amcehrphmotypesmaodal,1995;Changetal., 2001) as showninTablel andcanbereoognizedmorphologicallyas shownin Fig 3. BefomshrdyhgflreerqxessionofmaspmmflresecdlsTypeImxlTypeHHBECwae nmnmncytodrmfimflystanwdwimcytokaafinceflmmkasmrecmfinnflrdrcdltypes Asshownin arepresartative figure (Fig4), Type I HBEC ispositive forcytokeratin18,butmtcytoka'atin 14, whereasTypeIIHBECispositiveforcytoka'atin 14,butnotcytokeratinl8. 28 figure3 MorphologissonypeIflop)andTypeII(Bottom)HBECsofHME30 29 Cytokeratin 1 4 TI I HBEC T. [I HBEC Cytokeratin 18 “gum Expressbnonymkeratinl4andl8in'I‘ypeIand'I‘ypeIIHBECs TopRight: Cytokeralinl4Expre$ioninTypeIHME30 BounmRight: CymkmtinrsExpmaoninrypeIHMEso Toplcfi: Cytolmtin14ExprsssioninTypeIIHlv1E30 TopRight: Cytokeratinl8ExpressioninTypeIIIm’IE30 30 ToammeflreaqxemimmflcdhflarmhmfimofmaspmpotdnofflreZtypesofHBEC, irrnnmocytodremicalstairfingsofthesecellswerecaniedout. AsslnwninFigS,maspinexpression wasnotobsa‘vedinTypeII~IBEC,butwasclearlyseeninflrecytoplasmonypeHHBEC. AllTypeI andTypeHwflsfiomflretwodifi‘a'entI-IBECstestedaiMEwand31)similarlyslmwedthatthe maspinwasexprsssedinTypeIII-IBEC (Fig 5a, 5b),butnotinTypeII-IBEC (Fig 5a, 5b). chmfinnafimweflanbldmflyfiswasdmcmfiedommmflwmmmofmaqfin inTypeIarxlTypelIHBECderivedfiorntIneedifi‘a'entinfividuals Theresultsslnwflratmaspin(42 kD)washighlyeaq)ressedinTypeIIHBECs,lmtnotinTypeIHBECs(Fig 6). Althoughthelevelsof equsimmyvmymrmgdiflaancdlaflmflmommawngoppodmmessimofmaspinm TypeIarxlTypeIIHBECwasoonsistentintheseoellarlum 31 Typel .- Type“ - - figureSa MaspinExprssu’onbyImnnmostaininginT‘ypeIflofiandTypeIl (Bottom)HBECofHME30 'I‘hephaseconn'astirnagssofcellsareshownontheright. 32 FigureSb MaspinExpressionbyImrmmostnininginTypeIflop)andTypeII (Bottom)HBECofHME3l Thephmecontrmtimagesofoellsareshownonflreright. Lane 1 2 3 4 5 6 Maspin 1‘ M W Actin MR6 WestemetAnalysisofMasp’mExpresdonhTypeIandTypeII HBECofHME29(hnesl,2),HIVIIBO(hnes3,4)andHIVIE31(IanssS, 6) 34 Expreesionofmasphrinr'nmnansformedHBECandcanceroefllines A series ofin vitro immortalized and neoplastically transformed Type I HBEC lines (Ml38V1, Ml3SV1R2, Ml3SV1R2Nl) weredraracterized formaspin expression. As shown in Fig. 7, all these Type I HBEC neoplastically transformed at different stages (immortal Ml3SV1, weakly tumorigenic M13SV1R2 and highly tumorigenic M13SV1R2N1) were negative in maspin expression by irmnmncytodremicalsuxliessimilartononnalTypeIHBEC. Asexpected,thebreastcanoercelllines, MCF-7 (ER+) and MDA-MB-231 (ER-) were negtive in maspin expression (Fig 8) hrwestarrblotarralySisafltheinviuouansfonnedTypeIHBEClmesCLe immortalMlBSVl,weakly tumorigenic M13SV1R2 and highly trnnorigmic M13SV1R2N1) (Kang et al., 1998) as well as the MCF-7,MDA-MB-231de47Dbreastcmoa'cdlhnesweelbmdrutoexprelsflrenmspinpotein (Fig 9). 35 t :‘i FIgure7 MaspinExpreesionofInvitmTransfondeypeIHBECLinesat Difi'erentStageshy' ‘ ' ' "mt“ Thephasecontrastimagesofoelkareshownonflreright. Figure 8 Maspin Expression in Breast Carcinoma Cell Lines, MCF-7 (mp), MDA-MB-231 (bottom) by Immunostaining 'I‘hephaseoontrastimagesofwlkareshownontheright. 37 Lane 1 2 3 4 5 6 7 8 Maspin Actin h i I b» a fig art a r g .3 ‘3' Figure 9 Western Blot Analysis of Maspin Expression in normal HBEC, in Vitro Immortalized or Neoplastically Transformed Type I HBEC and Breast Carcinoma Cell Lines Lane 1: TypeIHME 30 Lane 5: M13SV1R2N1 Lane 2: Type II HME 30 Lane 6: MCF7 Lane 3: Ml3SV1 Lane 7: MDA-MB-231 Lane 4: M13SV1R22 Lane 8: T47D 38 DISCUSSION This study using immunocytochemical staining and Western blot analysis has demonstrated that the human breast epithelial cell type (Type I HBEC) with stem cell characteristics did not express maspin, whereas Type II HBEC with basal epithelial phenotypes expressed maspin. Furthermore, a series of step-wise neoplastically transformed cell lines derived from a Type I HBEC (Kang et al., 1998), i.e. immortal M 13SV1, weakly tumorigenic M 13SV1R2 and highly tumorigenic M 13SV1R2N1, did not show maspin expression. Previously, normal human breast epithelial cells (myoepithelial or basal epithelial) were shown to express maspin (Zou et al., 1994; Mass et al., 2001a; Maass et al., 2001b) whereas the majority of human breast cancer cell lines including MCF-7 and MDA-MB-231 did not show maspin expression (6/7 or 6/8) (Zou et al., 1994; Mass et al., 2002). Our experiment also confirmed that these two well- known breast cancer cell lines, MCF-7 and MDA-MB-231, did not express maspin. Therefore, the results are not consistent with the idea that maspin is expressed in all normal breast epithelial cells and that the expression of this serine proteinase inhibitor is down-regulated or mutated during tumor progression (Maass et al., 2001b). These results have several implications. First, similar to Cx 26, a-6 integrin and Oct-4 (Lee et al., 1991; Sager et al., 1993; Trosko et al., 2004) maspin is a breast epithelial stem cell marker whose expression is preserved from stem cells to tumorigenic cells as shown in the different Type I HBEC lines transformed at different stages. Although maspin has been clearly shown to possess tumor-suppressing activity (Zou et al., 1994), its down regulation in breast cancer cell lines (Zou et al., 1994; Maass et al., 2001a) or loss of 39 expression in tumor tissues, in contrast to cells in normal tissues such as myoepithelial cells (Maass et al., 2001b), does not mean a real change has occurred in gene expression from the target cells (i.e.Type I HBEC) to tumor cells. The results provide an alternative origin of a tumor suppressor gene and an additional mechanism explaining how stem cells could be target cells for carcinogenesis. Second, since maspin disappears when Type I HBECs differentiate into other cell types, maspin expression might be used to screen for chemopreventive agents. Indeed, sphingoid bases (sphinosine and sphinganine), which are potential chemopreventive agents, have been shown to induce the differentiation of Type I HBEC into Type II HBEC with the concomitant emergence of maspin expression in Type II HBEC (Ahn, 2003). The effect of this type of agent could reduce the target breast epithelial stem cells for carcinogenesis. Thirdly, since down-regulation of maspin could be a marker for breast cancer and its target precursor cell, the non-expression of this gene is expected to be early in precancerous cells, i.e., ductal carcinoma in situ. This gene could be used as a marker for early detection of precancerous cells. When Type I HBECs differentiate into Type II HBEC, there is a wholesale change in the expression of may genes and functions {Kao, Nomata, et al. 1995 24865 /id}{Chang, Sun, et al. 2001 19479 lid} {Chang, Tsai, et al. 2004 31265 /id} {Chang, Sun, et al. 2001 19479 lid} {Chang, Olson, et al. 2004 31314 /id}. These conversions are clearly due to 40 epi genetic changes during cell differentiation. For maspin, the re-expression of maspin in maspin-negative breast cancer lines (MCF-7, T-47D and MDA-MB-23 1) can be effected by treatment with 5-aza-2’deoxycytidine and/or trichostatin A, indicating that DNA methylation and/or histone deacetylation is/are partially responsible for the silencing of the maspin gene in breast cancer cells (Futscher et al., 2002). Furthermore, it has been shown that, in normal cells expressing maspin, the maspin promoter is unmethylated and the promoter region has acetylated histones (Maass et al., 2002; Futscher et al., 2002). In contrast, normal cells that do not express maspin have a completely methylated maspin promoter with hypoacetylated histones (Maass et al., 2002). These mechanisms are very likely to be involved in the silencing of maspin expression in Type I HBEC and can be examined in future studies. Bemards and Weinberg (Bemards and Weinberg, 2002) hypothesized that the metastatic behavior of cancer cells seems to be determined relatively early in tumorigenesis. They suggest that a subset of the mutant alleles acquired by incipient tmnor cells early in tumorigenesis confer not only the selected replicative advantage, but also, later in tumorigenesis, the proclivity to metastasize. Since maspin has been shown not only to inhibit the proteinase activity but also to reduce tumor growth through a combination of reduced angiogenesis and increased apoptosis (Shi et al., 2003), it is a gene fitting that description. However, the silencing of maspin in tumor cells might not be due to mutation or epigenetic alteration dming tumor progression, but rather due to continuous expression of a target stem cell phenotype. 41 Reference List Ahn, E. H. (2003). Chemopreventative and chemotherapeutic mechanisms of sphingolipid metabolites in human colon cancer cells and breast stem, normal and tumorigenic cells. Michigan State University. Akiyama,S.K., Olden,K., and Yamada KM. (1996). Fibronectin and integrins in invasion and metastasis. Cancer and metastasis reviews 14, 173-189. Alison,M., Golding,M., Lalani,E.N., Nagy,P., Thorgeirsson,S., and Sarraf,C. (1997). Wholesale hepatocytic differentiation in the rat fi'om ductular oval cells, the progeny of biliary stem cells. J. Hepatol. 26, 343-352. Arteaga,C.L. and Moses,H.L. (1996). TGF-beta in mammary development and neoplasia. J. Mammary. Gland. Biol. Neoplasia. 1, 327-329. Barker,K.T., Jackson,L.E., and Crompton,M.R. (1997). BRK tyrosine kinase expression in a high proportion of human breast carcinomas. Oncogene 15, 799-805. Barsky,S.H., Stemlicht,M.D., Safarians,S., Nguyen,M., Chin,K., Stewart,S.D., Hiti,A.L., and Gray,J.W. (1997). Evidence of a dominant transcriptional pathway which regulates an undifferentiated and complete metastatic phenotype. Oncogene 15, 2077-2091. Bemards,R. and Weinberg,R.A. (2002). A progression puzzle. Nature 418, 823. Chan,D., Goate,A., and Puck,T.T. (1989). Involvement of vimentin in the reverse transformation reaction. Proc. Natl. Acad. Sci. U. S. A 86, 2747-2751. Chang,C.C., Sun,W., Cruz,A., Saitoh,M., Tai,M.H., and Trosko,J.E. (2001). A human breast epithelial cell type with stem cell characteristics as target cells for carcinogenesis. Radiation Research 155, 201-207. Chang,C.C., Trosko,J.E., E1-Fou1y,M.H., Gibson,D.R., and D'Ambrosio,S.M. (1987). Contact insensitivity of a subpopulation of normal human fetal kidney epithelial cells and of human carcinoma cell lines. Cancer Res. 47, 1634-1645. Chang,C.C., Tsai,].L., Kuo,I(.K., and Wang,K.H. (2004). Expression of Oct-4, alpha fetoprotein and vimentin, and lack of gap junctional intercellular communication as common phenotypes for human adult liver stem cells and hepatoma cells. Abstract for Proc. Am. Assoc. Cancer Res. 45, 642. Dickson,R.B. (1994). Molecular basis of breast cancer. In The molecular basis of cancer, Mendelsohon, ed. (Philadelphia: W.B.Sunders Co.), pp. 358-384. Domann,F.E., Rice,J.C., Hendrix,M.J., and Futscher,B.W. (2000). Epigenetic silencing of maspin gene expression in human breast cancers. Int. J. Cancer 85, 805-810. 42 F edi,P. (1997). Growth factors. In Cancer Medicine, J .F.Holland, ed. (Baltimore: Williams and Wilkins), pp. 41-64. Foley,K.P. and Eisenman,R.N. (1999). Two MAD tails: what the recent knockouts of Madl and Mxil tell us about the MYC/MAX/MAD network. Biochim. Biophys. Acta 1423, M37-M47. Futscher,B.W., Oshiro,M.M., Wozniak,R.J., Holtan,N., Hanigan,C.L., Duan,H., and Domann,F.E. (2002). Role for DNA methylation in the control of cell type specific maspin expression. Nat. Genet. 31, 175-179. Ge, R. and Stanpfer, M and Reis M (2004). Endogenous transforming growth factor beta signalling and growth suppression is lost during mammary epithelial cell transformation. Proc. Am. Assoc. Cancer Res. 45, 811. Grueterich,M. and Tseng,S.C.G. (2002). Human limbal progenitor cells expanded on intact amniotic membrane ex vivo. Arch. Ophthalmol. 120, 783-790. Hahm, H A. (1998). Apoptosis in the mammary gland and breast cancer. Davidson and NE. Endocrine-Related Cancer 5, 199-211. Hanahan,D. and Folkman,J. (1996). Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis. Cell 86, 353-364. Hanahan,D. and Weinberg,R.A. (2000). The hallmarks of cancer. Cell 100, 57-70. Hayflick,L. (1997). Mortality and immortality at the cellular level. A review. Biochemistry (Mose. ) 62, 1 180-1 190. Herbert,B.S., Wright,W.E., and Shay,J.W. (2002). p16(INK4a) inactivation is not required to irnmortalize human mammary epithelial cells. Oncogene 21, 7897-7900. Hiyama,K., Hirai,Y., Kyoizumi,S., Akiyarna,M., Hiyama,E., Piatyszek,M.A., Shay,J.W., Ishioka,S., and Yamakido,M. (1995). Activation of telomerase in human lymphocytes and hematopoietic progenitor cells. .1 . hnmunol. 155, 3711-3715. Jacobs,C. and Rubsamen,H. (1983). Expression of pp600-src protein kinase in adult and fetal human tissue: high activities in some sarcomas and mammary carcinomas. Cancer Res. 43, 1696-1702. Jiang,N., Meng,Y., Zhang,S., Mensah-Osman,E., and Sheng,S. (2002). Maspin sensitizes breast carcinoma cells to induced apoptosis. Oncogene 21, 4089-4098. J in, L. (1996). Expression of scatter factor and c-met receptor in benign and malignant breast tissue. Fuchs, A. and Schnitt S.J.Yao, Y. Cancer (Phila) 79, 749-760. 43 Kang,K.S., Chang,C.C., and Trosko,J.E. (1998). Modulation of gap junctional intercellular communication during human breast stem cell differentiation and immortalization. In Gap Junctions, R.Werner, ed. 108 Press), pp. 347-351. Kang,K.S., Morita,I., Cruz,A., Jeon,Y.J., Trosko,J.E., and Chang,C.C. (1997). Expression of estrogen receptors in a normal human breast epithelial cell type with luminal and stem cell characteristics and its neoplastically transformed cell lines. Carcinogenesis 18, 251- 257. Kao,C.Y., Nomata,K., Oakley,C.S., Welsch,C.W., and Chang,C.C. (1995). Two types of normal human breast epithelial cells derived from reduction mammoplasty: phenotypic characterization and response to SV40 transfection. Carcinogenesis 16, 531-538. Kerr,J.F., Wyllie,A.H., and Currie,A.R. (1972). Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. Br. J. Cancer 26, 239-257. Kiyono,T., Foster,S.A., Koop,J.I., McDouga11,J.K., Galloway,D.A., and Klingelhutz,A.J. (1998). Both Rb/pl6INK4a inactivation and telomerase activity are required to immortalize human epithelial cells. Nature 396, 84-88. Kochetkova,M., McKenzie,O.L., Bais,A.J., Martin,J.M., Secker,G.A., Seshadri,R., Powell,J.A., Hinze,S.J., Gardner,A.E., Spendlove,H.E., O'Callaghan,N.J., Cleton- J ansen,A.M., Comelisse,C., Whitmore,S.A., Crawford,J., Kremmidiotis,G., Sutherland,G.R., and Callen,D.F. (2002). CBFA2T3 (MTGl6) is a putative breast tumor suppressor gene from the breast cancer loss of heterozygosity region at 16q24.3. Cancer Res. 62, 4599-4604. Kraus,M.H., Popescu,N.C., Amsbaugh,S.C., and King,C.R. (1987). Overexpression of the EGF receptor-related proto-oncogene erbB-2 in human mammary tumor cell lines by different molecular mechanisms. EMBO J. 6, 605-610. Kyo,S., Takakura,M., Kanaya,T., Zhuo,W., Fujimoto,K., Nishio,Y., Orimo,A., and Inoue,M. (1999). Estrogen activates telomerase. Cancer Res. 59, 5917-5921. Lee,S.W., Tomasetto,C., and Sager,R. (1991). Positive selection of candidate turnor- suppressor genes by subtractive hybridization. Proc. Natl. Acad. Sci. U. S. A. 88, 2825- 2829. Lele,S.M., Graves,K., and Gatalica,Z. (2000). Immunohistochemical detection of maspin is a useful adjunct in distinguishing radial sclerosing lesion from tubular carcinoma of the breast. Appl. Irnmunohistochem. Mol. Morphol. 8, 32-36. Li,J.J., Colbum,N.H., and Oberley,L.W. (1998). Maspin gene expression in tumor suppression induced by overexpressing manganese-containing superoxide dismutase cDNA in human breast cancer cells. Carcinogenesis 19, 833-839. Lin,T.M., Tsai,].L., Tai,M.H., Lin,S.D., Lai,C.S., Hsieh,Y.L., Cheng,K.I., and Chang,C.C. (2004). A new cell culture method to grow human mesenchymal 44 stem/progenitor cells with high proliferation and differentiation potential from adipose tissues. In Int.Soc.Stem Cell Res.Meeting Abstract, p. 99. Lippman,M.E. and and Allegra,J.C. (1980). Quantative estrogen receptor analysis: The response to endocrine and cytotoxic chemotherapy in human breast cancer and the disease-free interval. Cancer 46, 2829-2834. Lippman,S.M., Alberts,D.S., Slymen,D.J., Weiner,S., Aristizabal,S.A., Luditch,A., Davis,J.R., and Surwit,E.A. (1988). Second-look laparotomy in epithelial ovarian carcinoma. Prognostic factors associated with survival duration. Cancer 61, 2571-2577. Luttrell,D.K., Lee,A., Lansing,T.J., Crosby,R.M., Jung,K.D., Willard,D., Luther,M., Rodriguez,M., Berman,J., and Gilmer,T.M. (1994). Involvement of pp60c-src with two major signaling pathways in human breast cancer. Proc. Natl. Acad. Sci. U. S. A 91, 83- 87. Maass,N., Biallek,M., Rosel,F., Schem,C., Ohike,N., Zhang,M., Jonat,W., and Nagasaki,K. (2002). Hyperrnethylation and histone deacetylation lead to silencing of the maspin gene in human breast cancer. Biochem. Biophys. Res. Commun. 297, 125-128. Maass,N., Hojo,T., Ueding,M., Luttges,J., Kloppel,G., Jonat,W., and Nagasaki,K. (2001 a). Expression of the tumor suppressor gene Maspin in human pancreatic cancers. Clin. Cancer Res. 7, 812-817. Maass,N., Hojo,T., Zhang,M., Sager,R., Jonat,W., and Nagasaki,K. (2000). Maspin--a novel protease inhibitor with tumor-suppressing activity in breast cancer. Acta Oncol. 39, 931-934. Maass,N., Teffner,M., Rose1,F ., Pawaresch,R., Jonat,W., Nagasaki,K., and Rudolph,P. (2001b). Decline in the expression of the serine proteinase inhibitor maspin is associated with tumour progression in ductal carcinomas of the breast. J. Pathol. 195, 321-326. Manni,A., Arafah,B., and Pearson,O.H. (1980). Estrogen and progesterone receptors in the prediction of response of breast cancer to endocrine therapy. Cancer 46, 283 8-2841 . Markert,C.L. (1968). Neoplasia: a disease of cell differentiation. Cancer Res. 28, 1908- 1914. Martin,K.J., Kritzrnan,B.M., Price,L.M., Koh,B., Kwan,C.P., Zhang,X., MacKay,A., O'Hare,M.J., Kaelin,C.M., Mutter,G.L., Pardee,A.B., and Sager,R. (2000). Linking gene expression patterns to therapeutic groups in breast cancer. Cancer Res. 60, 2232-2238. Mathon,N.F., Malcolm,D.S., Harrisingh,M.C., Cheng,L., and Lloyd,A.C. (2001). Lack of replicative senescence in normal rodent glia cells. Science 291, 868-871. Matic,M., Evans,W.H., Brink,P.R., and Simon,M. (2002). Epidermal stem cells do not communicate through gap junctions. J. Invest. Dermatol. 118, 110-116. 45 Mitche11,P.J., Barker,K.T., Martindale,J.E., Kamalati,T., Lowe,P.N., Page,M.J., Gusterson,B.A., and Crompton,M.R. (1994). Cloning and characterisation of cDNAs encoding a novel non-receptor tyrosine kinase, brk, expressed in human breast tumours. Oncogene 9, 2383-2390. Moses,H.L., Yang,E.Y., and Pietenpol,J.A. (1990). TGF-beta stimulation and inhibition of cell proliferation: new mechanistic insights. Cell 63, 245-247. Nakatani,K., Thompson,D.A., Barthe1,A., Sakaue,H., Liu,W., Weigel,R.J., and Roth,R.A. (1999). Up-regulation of Akt3 in estrogen receptor-deficient breast cancers and androgen-independent prostate cancer lines. J. Biol. Chem. 274, 21528-21532. Nonet,G.H., Stampfer,M.R., Chin,K., Gray,J.W., Collins,C.C., and Yaswen,P. (2001). The ZN F 217 gene amplified in breast cancers promotes immortalization of human mammary epithelial cells. Cancer Res. 61, 1250-1254. Pemberton,P.A., Wong,D.T., Gibson,H.L., Kiefer,M.C., Fitzpatrick,P.A., Sager,R., and Barr,P.J. (1995). The tumor suppressor maspin does not undergo the stressed to relaxed transition or inhibit trypsin-like serine proteases. Evidence that maspin is not a protease inhibitory serpin. J. Biol. Chem. 270, 15832-15837. Porter,D.A., Krop,I.E., Nasser,S., Sgroi,D., Kaelin,C.M., Marks,J.R., Riggins,G., and Polyak,K. (2001). A SAGE (serial analysis of gene expression) view of breast tumor progression. Cancer Res. 61, 5697-5702. Potter,V.R. (1978). Phenotypic diversity in experimental hepatomas: The concept of partially blocked ontogeny. Br. .1. Cancer 38, 1-23. Reddy,D.E., Keck,C.L., Popescu,N., Athwa1,R.S., and Kaur,G.P. (2000). Identification of a YAC from 16q24 carrying a senescence gene for breast cancer cells. Oncogene 19, 217- 222. Reddy,D.E., Sandhu,A.K., DeRiel,J.K., Athwa1,R.S., and Kaur,G.P. (1999). Identification of a gene at 16q24.3 that restores cellular senescence in immortal mammary tumor cells. Oncogene 18, 5100-017. Reis-F i1ho,J .S., Milanezi,F., Silva,P., and Schmitt,F.C. (2001). Maspin expression in myoepithelial tumors of the breast. Pathol. Res. Pract. 197, 817-821. Russo,I.H. and Russo,J. (1996). Mammary gland neoplasia in long-terrn rodent studies. Environ. Health Perspect. 104, 938-967. Russo,I.H. and Russo,J. (1998). Role of hormones in mammary cancer initiation and progression. J. Mammary. Gland. Biol. Neoplasia 3, 49-61. Russo,J. and Russo,I.H. (1997). Role of differentiation in the pathogenesis and prevention of breast cancer. Endocrine Related Cancer 4, 7-21. 46 Sager,R., Anisowicz,A., Neveu,M., Liang,P., and Sotiropoulou,G. (1993). Identification by differential display of alpha 6 integin as a candidate tumor suppressor gene. FASEB J. 7, 964-970. Sager,R., Sheng,S., Anisowicz,A., Sotiropoulou,G., Zou,Z., Stenman,G., Swisshelm,K., Chen,Z., Hendrix,M.J., Pemberton,P., and et,a. (1994). RNA genetics of breast cancer: maspin as paradign. Cold Spring Harb. Syrnp. Quant. Biol. 59, 537-546. Sawyers,C.L., Denny,C.T., and Witte,O.N. (1991). Leukemia and the disruption of normal hematopoiesis. Cell 64, 337-350. Schneider,S.S., Schick,C., Fish,K.E., Miller,E., Pena,J.C., Treter,S.D., Hui,S.M., and Silverman,G.A. (1995). A serine proteinase inhibitor locus at 18q21.3 contains a tandem duplication of the human squamous cell carcinoma antigen gene. Proc. Natl. Acad. Sci. U. S. A 92, 3147-3151. Schwarze,P.E., Pettersen,E.O., Shoaib,M.C., and Seglen,P.O. (1984). Emergence of a population of small, diploid hepatocytes during hepatocarcinogenesis. Carcinogenesis 5, 1267-1275. Sell,S. (1993). Cellular origin of cancer: dedifferentiation or stem cell maturation arrest? Environ. Health Perspect. 101 Suppl 5, 15-26. Shao, Z. M., Nguyen, M., Alpaugh, M. L., O'Connell, J. T., and Barsky, S. H. (1998). The human myoepithelial cell exerts antiproliferative effects on breast carcinoma cells characterized by p21(W AF l/CIPI) induction, G(2)/M arrest, and apoptosis. Exp.Cell Res. 241[2], 394-403. Sheng,S., Carey,J., Seftor,E.A., Dias,L., Hendrix,M.J., and Sager,R. (1996). Maspin acts at the cell membrane to inhibit invasion and motility of mammary and prostatic cancer cells. Proc. Natl. Acad. Sci. U. S. A 93, 1 1669-11674. Sheng,S., Truong,B., Fredrickson,D., Wu,R., Pardee,A.B., and Sager,R. (1998). Tissue- type plasminogen activator is a target of the tumor suppressor gene maspin. Proc. Natl. Acad. Sci. U. S. A 95, 499-504. Shi,H.Y., Zhang,W., Liang,R., Abraham,S., Kittrell,F.S., Medina,D., and Zhang,M. (2001). Blocking tumor growth, invasion, and metastasis by maspin in a syngeneic breast cancer model. Cancer Res. 61 , 6945-6951. Shi,H.Y., Zhang,W., Liang,R., Kittrell,F., Templeton,N.S., Medina,D., and Zhang,M. (2003). Modeling human breast cancer metastasis in mice: maspin as a paradign. Histol. Histopathol. 18, 201-206. Slamon,D.J., Clark,G.M., Wong,S.G., Levin,W.J., Ullrich,A., and McGuire,W.L. (1987). Human breast cancer: correlation of relapse and survival with amplification of the HER- 2/neu oncogene. Science 235, 177-182. 47 Stemlicht,M.D. and Barsky,S.H. (1997). The myoepithelial defense: a host defense against cancer. Med. Hypotheses 48, 37-46. Stemlicht,M.D., Kedeshian,P., Shao,Z.M., Safarians,S., and Barsky,S.H. (1997). The human myoepithelial cell is a natural tumor suppressor. Clin. Cancer Res. 3, 1949-1958. Streuli,C.H. (2002). Maspin is a tumour suppressor that inhibits breast cancer tumour metastasis in vivo. Breast Cancer Res. 4, 137-140. Styles,J., Elliott,B.M., Lefevre,P.A., Robinson,M., Pritchard,N., Hart,D., and Ashby,J. (1985). Irreversible depression in the ratio of tetraploidzdiploid liver nuclei in rats treated with 3'-methyl-4-dimethy1aminoazobenzene(3'M). Carcinogenesis 6, 21-28. Sun,W., Kang,K.S., Morita,I., Trosko,J.E., and Chang,C.C. ( 1999). High susceptibility of a human breast epithelial cell type with stem cell characteristics to telomerase activation and immortalization. Cancer Res. 59, 6118-6123. Tang,D.G., Tokumoto,Y.M., Apperly,J.A., Lloyd,A.C., and Raff,M.C. (2001). Lack of replicative senescence in cultured rat oligodentrocyte precursor cells. Science 291 , 872- 875. Thomson,J.A., Itskovitz,E.J., Shapiro,S.S., Waknitz,M.A., Swiergiel,J.J., Marshall,V.S., and J ones,J .M. (1998). Embryonic stem cell lines derived from human blastocysts. Science 282, 1145-1147. Trosko,J.E. (2003). The role of stem cells and gap junctional intercellular communication in carcinogenesis. J. Biochem. Mol. Biol. 36, 43-48. Trosko,J.E., Chang,C.C., Wilson,M.R., Upham,B., Hayashi,T., and Wade,M. (2000). Gap junctions and the regulation of cellular functions of stem cells during development and differentiation. Methods 20, 245-264. Trosko,J.E., Tai,M.H., Chang,C.C., Olson,L.K., Lin,T.M., and Madhukar,B.V. (2004). Expression of Oct-4 and lack of gap junctional intercellular communication as "hallmarks" for human adult stem cells and cancer cells. Proc. Am. Assoc. Cancer Res. 45, 643. Tsai,R.Y. and McKay,R.D. (2002). A nucleolar mechanism controlling cell proliferation in stem cells and cancer cells. Genes Dev. 16, 2991-3003. Tuck,A.B., Park,M., Sterns,E.E., Boag,A., and Elliott,B.E. (1996). Coexpression of hepatocyte gowth factor and receptor (Met) in human breast carcinoma. Am. J. Pathol. 148, 225-232. Van de Vijver,M.J. and Nusse,R. (1991). The molecular biology of breast cancer. Biochim. Biophys. Acta 1072, 33-50. 48 Varmus,H. and Weinberg,R.A. (1993). Genes and the biology of cancer. W.H. Freeman and Co.), pp. 38-39. Wang,Y., Selden,C., Famaud,S., Calnan,D., and Hodgson,H.J. (1994). Hepatocyte gowth factor (HGF/SF) is expressed in human epithelial cells during embryonic development; studies by in situ hybridisation and northern blot analysis. J. Anat. 185 ( Pt 3), 543-551. Watson,P.H., Pon,R.T., and Shiu,R.P. (1991). Inhibition of c-myc expression by phosphorothioate antisense oligonucleotide identifies a critical role for c-myc in the gowth of human breast cancer. Cancer Res. 51, 3996-4000. Wazer,D.E., Liu,X.L., Chu,Q., Gao,Q., and Band,V. (1995). Irnmortalization of distinct human mammary epithelial cell types by human papilloma virus 16 E6 or E7. Proc. Natl. Acad. Sci U. S. A 92, 3687-3691. Yamasaki,H., Hollstein,M., Mesni1,M., Martel,N., and Aguelon,A.M. (1987). Selective lack of intercellular communication between transformed and nontransformed cells as a common property of chemical and oncogene transformation of BALB/c3T3 cells. Cancer Res. 47, 5658-5664. Zhang,M., Shi,Y., Magit,D., Furth,P.A., and Sager,R. (2000a). Reduced mammary tumor progession in WAP-TAg/WAP-maspin bitransgenic mice. Oncogene 19, 6053-6058. Zhang,M., Volpert,O., Shi,Y.H., and Bouck,N. (2000b). Maspin is an angiogenesis inhibitor. Nat. Med. 6, 196-199. Zou,Z., Anisowicz,A., Hendrix,M.J., Thor,A., Neveu,M., Sheng,S., Rafidi,K., Seftor,E., and Sager,R. (1994). Maspin, a serpin with tumor-suppressing activity in human mammary epithelial cells. Science 263, 526-529. Zou,Z., Gao,C., Nagaich,A.K., Connell,T., Saito,S., Mou1,J.W., Seth,P., Appella,E., and Srivastava,S. (2000). p53 regulates the expression of the tumor suppressor gene maspin. J. Biol. Chem. 275, 6051-6054. 49 lliliiliilijiil111111